US20200080154A1 - Methods of diagnosing and treating alzheimer's disease - Google Patents
Methods of diagnosing and treating alzheimer's disease Download PDFInfo
- Publication number
- US20200080154A1 US20200080154A1 US16/610,293 US201816610293A US2020080154A1 US 20200080154 A1 US20200080154 A1 US 20200080154A1 US 201816610293 A US201816610293 A US 201816610293A US 2020080154 A1 US2020080154 A1 US 2020080154A1
- Authority
- US
- United States
- Prior art keywords
- exon
- app
- classical variant
- classical
- probe
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Pending
Links
- 238000000034 method Methods 0.000 title claims abstract description 254
- 208000024827 Alzheimer disease Diseases 0.000 title claims abstract description 135
- 230000014509 gene expression Effects 0.000 claims abstract description 274
- 230000000694 effects Effects 0.000 claims abstract description 206
- 108090000623 proteins and genes Proteins 0.000 claims abstract description 125
- 239000000523 sample Substances 0.000 claims description 265
- 108091032973 (ribonucleotides)n+m Proteins 0.000 claims description 224
- 108020004414 DNA Proteins 0.000 claims description 186
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 claims description 161
- 125000003729 nucleotide group Chemical group 0.000 claims description 150
- 239000002773 nucleotide Substances 0.000 claims description 144
- 101150031224 app gene Proteins 0.000 claims description 132
- 102000013455 Amyloid beta-Peptides Human genes 0.000 claims description 127
- 108010090849 Amyloid beta-Peptides Proteins 0.000 claims description 127
- 239000003795 chemical substances by application Substances 0.000 claims description 114
- 102000004169 proteins and genes Human genes 0.000 claims description 101
- 239000012472 biological sample Substances 0.000 claims description 99
- 238000009825 accumulation Methods 0.000 claims description 96
- 201000010099 disease Diseases 0.000 claims description 81
- 208000035475 disorder Diseases 0.000 claims description 80
- 239000000074 antisense oligonucleotide Substances 0.000 claims description 78
- 238000012230 antisense oligonucleotides Methods 0.000 claims description 78
- 108091034117 Oligonucleotide Proteins 0.000 claims description 74
- 150000001413 amino acids Chemical group 0.000 claims description 72
- 239000004055 small Interfering RNA Substances 0.000 claims description 66
- 238000007671 third-generation sequencing Methods 0.000 claims description 62
- 238000007901 in situ hybridization Methods 0.000 claims description 53
- 239000012634 fragment Substances 0.000 claims description 44
- 102220006209 rs193922916 Human genes 0.000 claims description 37
- 102200131746 rs63750066 Human genes 0.000 claims description 37
- 102200131813 rs63750264 Human genes 0.000 claims description 37
- 102200131815 rs63750264 Human genes 0.000 claims description 37
- 102200131764 rs63750734 Human genes 0.000 claims description 37
- 102220032732 rs63750868 Human genes 0.000 claims description 37
- 102200131757 rs63750973 Human genes 0.000 claims description 37
- 102200131811 rs63751122 Human genes 0.000 claims description 37
- 102220263566 rs772168426 Human genes 0.000 claims description 37
- 108020004459 Small interfering RNA Proteins 0.000 claims description 36
- 108700011259 MicroRNAs Proteins 0.000 claims description 35
- 239000002679 microRNA Substances 0.000 claims description 35
- 108010079054 Amyloid beta-Protein Precursor Proteins 0.000 claims description 34
- 108090000765 processed proteins & peptides Proteins 0.000 claims description 34
- 108091027967 Small hairpin RNA Proteins 0.000 claims description 30
- 238000010379 pull-down assay Methods 0.000 claims description 30
- 238000003753 real-time PCR Methods 0.000 claims description 29
- HOKKHZGPKSLGJE-GSVOUGTGSA-N N-Methyl-D-aspartic acid Chemical compound CN[C@@H](C(O)=O)CC(O)=O HOKKHZGPKSLGJE-GSVOUGTGSA-N 0.000 claims description 28
- 239000000427 antigen Substances 0.000 claims description 28
- 108091007433 antigens Proteins 0.000 claims description 28
- 102000036639 antigens Human genes 0.000 claims description 28
- 208000025688 early-onset autosomal dominant Alzheimer disease Diseases 0.000 claims description 27
- 208000015756 familial Alzheimer disease Diseases 0.000 claims description 27
- 239000002299 complementary DNA Substances 0.000 claims description 26
- 101710154606 Hemagglutinin Proteins 0.000 claims description 23
- 101710093908 Outer capsid protein VP4 Proteins 0.000 claims description 23
- 101710135467 Outer capsid protein sigma-1 Proteins 0.000 claims description 23
- 101710176177 Protein A56 Proteins 0.000 claims description 23
- 210000004369 blood Anatomy 0.000 claims description 23
- 239000008280 blood Substances 0.000 claims description 23
- 239000000185 hemagglutinin Substances 0.000 claims description 23
- 230000002452 interceptive effect Effects 0.000 claims description 21
- YBJHBAHKTGYVGT-ZKWXMUAHSA-N (+)-Biotin Chemical compound N1C(=O)N[C@@H]2[C@H](CCCCC(=O)O)SC[C@@H]21 YBJHBAHKTGYVGT-ZKWXMUAHSA-N 0.000 claims description 18
- 102000005720 Glutathione transferase Human genes 0.000 claims description 18
- 108010070675 Glutathione transferase Proteins 0.000 claims description 18
- 230000002285 radioactive effect Effects 0.000 claims description 18
- 150000003384 small molecules Chemical class 0.000 claims description 18
- 102000040430 polynucleotide Human genes 0.000 claims description 17
- 108091033319 polynucleotide Proteins 0.000 claims description 17
- 239000002157 polynucleotide Substances 0.000 claims description 17
- 210000001175 cerebrospinal fluid Anatomy 0.000 claims description 16
- 238000009396 hybridization Methods 0.000 claims description 16
- 108091023037 Aptamer Proteins 0.000 claims description 14
- 108020004638 Circular DNA Proteins 0.000 claims description 14
- BFMYDTVEBKDAKJ-UHFFFAOYSA-L disodium;(2',7'-dibromo-3',6'-dioxido-3-oxospiro[2-benzofuran-1,9'-xanthene]-4'-yl)mercury;hydrate Chemical compound O.[Na+].[Na+].O1C(=O)C2=CC=CC=C2C21C1=CC(Br)=C([O-])C([Hg])=C1OC1=C2C=C(Br)C([O-])=C1 BFMYDTVEBKDAKJ-UHFFFAOYSA-L 0.000 claims description 14
- ADEBPBSSDYVVLD-UHFFFAOYSA-N donepezil Chemical group O=C1C=2C=C(OC)C(OC)=CC=2CC1CC(CC1)CCN1CC1=CC=CC=C1 ADEBPBSSDYVVLD-UHFFFAOYSA-N 0.000 claims description 14
- ASUTZQLVASHGKV-JDFRZJQESA-N galanthamine Chemical compound O1C(=C23)C(OC)=CC=C2CN(C)CC[C@]23[C@@H]1C[C@@H](O)C=C2 ASUTZQLVASHGKV-JDFRZJQESA-N 0.000 claims description 14
- 239000000816 peptidomimetic Substances 0.000 claims description 14
- 102000004196 processed proteins & peptides Human genes 0.000 claims description 14
- 229940044551 receptor antagonist Drugs 0.000 claims description 14
- 239000002464 receptor antagonist Substances 0.000 claims description 14
- 229940122041 Cholinesterase inhibitor Drugs 0.000 claims description 13
- 239000000544 cholinesterase inhibitor Substances 0.000 claims description 13
- 238000000151 deposition Methods 0.000 claims description 13
- 230000008021 deposition Effects 0.000 claims description 13
- 229920001184 polypeptide Polymers 0.000 claims description 13
- 108091092566 Extrachromosomal DNA Proteins 0.000 claims description 12
- 230000001900 immune effect Effects 0.000 claims description 11
- 230000002441 reversible effect Effects 0.000 claims description 11
- AUTOLBMXDDTRRT-JGVFFNPUSA-N (4R,5S)-dethiobiotin Chemical compound C[C@@H]1NC(=O)N[C@@H]1CCCCCC(O)=O AUTOLBMXDDTRRT-JGVFFNPUSA-N 0.000 claims description 9
- HNDVDQJCIGZPNO-YFKPBYRVSA-N L-histidine Chemical compound OC(=O)[C@@H](N)CC1=CN=CN1 HNDVDQJCIGZPNO-YFKPBYRVSA-N 0.000 claims description 9
- 229960002685 biotin Drugs 0.000 claims description 9
- 235000020958 biotin Nutrition 0.000 claims description 9
- 239000011616 biotin Substances 0.000 claims description 9
- 239000003153 chemical reaction reagent Substances 0.000 claims description 9
- HNDVDQJCIGZPNO-UHFFFAOYSA-N histidine Natural products OC(=O)C(N)CC1=CN=CN1 HNDVDQJCIGZPNO-UHFFFAOYSA-N 0.000 claims description 9
- 229920002704 polyhistidine Polymers 0.000 claims description 9
- 229940099433 NMDA receptor antagonist Drugs 0.000 claims description 7
- XSVMFMHYUFZWBK-NSHDSACASA-N Rivastigmine Chemical compound CCN(C)C(=O)OC1=CC=CC([C@H](C)N(C)C)=C1 XSVMFMHYUFZWBK-NSHDSACASA-N 0.000 claims description 7
- 229950008995 aducanumab Drugs 0.000 claims description 7
- 229950001863 bapineuzumab Drugs 0.000 claims description 7
- 229950001954 crenezumab Drugs 0.000 claims description 7
- 229960003530 donepezil Drugs 0.000 claims description 7
- 229960003980 galantamine Drugs 0.000 claims description 7
- ASUTZQLVASHGKV-UHFFFAOYSA-N galanthamine hydrochloride Natural products O1C(=C23)C(OC)=CC=C2CN(C)CCC23C1CC(O)C=C2 ASUTZQLVASHGKV-UHFFFAOYSA-N 0.000 claims description 7
- 229950002508 gantenerumab Drugs 0.000 claims description 7
- BUGYDGFZZOZRHP-UHFFFAOYSA-N memantine Chemical group C1C(C2)CC3(C)CC1(C)CC2(N)C3 BUGYDGFZZOZRHP-UHFFFAOYSA-N 0.000 claims description 7
- 229960004640 memantine Drugs 0.000 claims description 7
- 239000003703 n methyl dextro aspartic acid receptor blocking agent Substances 0.000 claims description 7
- 229950003486 ponezumab Drugs 0.000 claims description 7
- 229960004136 rivastigmine Drugs 0.000 claims description 7
- 229950007874 solanezumab Drugs 0.000 claims description 7
- 238000000159 protein binding assay Methods 0.000 claims description 4
- 102000014303 Amyloid beta-Protein Precursor Human genes 0.000 claims 26
- 102100022704 Amyloid-beta precursor protein Human genes 0.000 abstract description 493
- DZHSAHHDTRWUTF-SIQRNXPUSA-N amyloid-beta polypeptide 42 Chemical compound C([C@@H](C(=O)N[C@@H](C)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC(O)=O)C(=O)N[C@H](C(=O)NCC(=O)N[C@@H](CO)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CCCCN)C(=O)NCC(=O)N[C@@H](C)C(=O)N[C@H](C(=O)N[C@@H]([C@@H](C)CC)C(=O)NCC(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCSC)C(=O)N[C@@H](C(C)C)C(=O)NCC(=O)NCC(=O)N[C@@H](C(C)C)C(=O)N[C@@H](C(C)C)C(=O)N[C@@H]([C@@H](C)CC)C(=O)N[C@@H](C)C(O)=O)[C@@H](C)CC)C(C)C)NC(=O)[C@H](CC=1C=CC=CC=1)NC(=O)[C@@H](NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CCCCN)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](CC=1N=CNC=1)NC(=O)[C@H](CC=1N=CNC=1)NC(=O)[C@@H](NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)CNC(=O)[C@H](CO)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CC=1N=CNC=1)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CC=1C=CC=CC=1)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](C)NC(=O)[C@@H](N)CC(O)=O)C(C)C)C(C)C)C1=CC=CC=C1 DZHSAHHDTRWUTF-SIQRNXPUSA-N 0.000 abstract description 485
- 101710137189 Amyloid-beta A4 protein Proteins 0.000 abstract description 483
- 101710151993 Amyloid-beta precursor protein Proteins 0.000 abstract description 483
- 235000018102 proteins Nutrition 0.000 description 89
- 235000001014 amino acid Nutrition 0.000 description 67
- 108020005544 Antisense RNA Proteins 0.000 description 55
- 239000003184 complementary RNA Substances 0.000 description 55
- 150000007523 nucleic acids Chemical class 0.000 description 42
- 108700024394 Exon Proteins 0.000 description 41
- 102000039446 nucleic acids Human genes 0.000 description 37
- 108020004707 nucleic acids Proteins 0.000 description 37
- 238000012163 sequencing technique Methods 0.000 description 30
- 239000003814 drug Substances 0.000 description 29
- 229940124597 therapeutic agent Drugs 0.000 description 29
- 108020004999 messenger RNA Proteins 0.000 description 27
- 238000013518 transcription Methods 0.000 description 25
- 230000035897 transcription Effects 0.000 description 25
- 230000000692 anti-sense effect Effects 0.000 description 24
- 210000004556 brain Anatomy 0.000 description 24
- 210000004027 cell Anatomy 0.000 description 24
- 210000004940 nucleus Anatomy 0.000 description 24
- 238000012360 testing method Methods 0.000 description 23
- 230000008859 change Effects 0.000 description 21
- 230000000295 complement effect Effects 0.000 description 20
- 239000012099 Alexa Fluor family Substances 0.000 description 19
- 230000033616 DNA repair Effects 0.000 description 18
- 108091028043 Nucleic acid sequence Proteins 0.000 description 18
- 229940024606 amino acid Drugs 0.000 description 18
- 238000012228 RNA interference-mediated gene silencing Methods 0.000 description 17
- 230000009368 gene silencing by RNA Effects 0.000 description 17
- 210000002569 neuron Anatomy 0.000 description 17
- 230000037361 pathway Effects 0.000 description 17
- 239000007850 fluorescent dye Substances 0.000 description 15
- 235000000346 sugar Nutrition 0.000 description 15
- PXHVJJICTQNCMI-UHFFFAOYSA-N Nickel Chemical compound [Ni] PXHVJJICTQNCMI-UHFFFAOYSA-N 0.000 description 14
- -1 PerCP Chemical compound 0.000 description 14
- 238000012217 deletion Methods 0.000 description 14
- 230000037430 deletion Effects 0.000 description 14
- RWSXRVCMGQZWBV-WDSKDSINSA-N glutathione Chemical compound OC(=O)[C@@H](N)CCC(=O)N[C@@H](CS)C(=O)NCC(O)=O RWSXRVCMGQZWBV-WDSKDSINSA-N 0.000 description 14
- 238000003752 polymerase chain reaction Methods 0.000 description 14
- 238000003757 reverse transcription PCR Methods 0.000 description 14
- 238000002965 ELISA Methods 0.000 description 12
- 238000001502 gel electrophoresis Methods 0.000 description 12
- 230000006780 non-homologous end joining Effects 0.000 description 12
- 238000000338 in vitro Methods 0.000 description 11
- 239000003112 inhibitor Substances 0.000 description 11
- 230000004048 modification Effects 0.000 description 11
- 238000012986 modification Methods 0.000 description 11
- 230000008707 rearrangement Effects 0.000 description 11
- 238000003556 assay Methods 0.000 description 10
- 238000003559 RNA-seq method Methods 0.000 description 9
- 102220429978 c.2061G>C Human genes 0.000 description 9
- 238000006243 chemical reaction Methods 0.000 description 9
- 102200010594 rs1554200371 Human genes 0.000 description 9
- 102200133529 rs1555995816 Human genes 0.000 description 9
- 102200131748 rs63749810 Human genes 0.000 description 9
- 102200131812 rs63749964 Human genes 0.000 description 9
- 102200131773 rs63750064 Human genes 0.000 description 9
- 102220248824 rs63750064 Human genes 0.000 description 9
- 102220032735 rs63750151 Human genes 0.000 description 9
- 102200131810 rs63750264 Human genes 0.000 description 9
- 102200131814 rs63750399 Human genes 0.000 description 9
- 102200131756 rs63750643 Human genes 0.000 description 9
- 102220032734 rs63750851 Human genes 0.000 description 9
- 102200131753 rs63751039 Human genes 0.000 description 9
- 108020005004 Guide RNA Proteins 0.000 description 8
- 108060003951 Immunoglobulin Proteins 0.000 description 8
- 238000002105 Southern blotting Methods 0.000 description 8
- 102000018358 immunoglobulin Human genes 0.000 description 8
- 230000005764 inhibitory process Effects 0.000 description 8
- 230000035772 mutation Effects 0.000 description 8
- 238000012216 screening Methods 0.000 description 8
- 241000894007 species Species 0.000 description 8
- 238000011282 treatment Methods 0.000 description 8
- 108020000948 Antisense Oligonucleotides Proteins 0.000 description 7
- 108090001008 Avidin Proteins 0.000 description 7
- 108010024636 Glutathione Proteins 0.000 description 7
- 108010021625 Immunoglobulin Fragments Proteins 0.000 description 7
- 102000008394 Immunoglobulin Fragments Human genes 0.000 description 7
- 108091092195 Intron Proteins 0.000 description 7
- 108010090804 Streptavidin Proteins 0.000 description 7
- 230000003321 amplification Effects 0.000 description 7
- 239000000975 dye Substances 0.000 description 7
- 229960003180 glutathione Drugs 0.000 description 7
- 230000006801 homologous recombination Effects 0.000 description 7
- 238000002744 homologous recombination Methods 0.000 description 7
- 238000003780 insertion Methods 0.000 description 7
- 230000037431 insertion Effects 0.000 description 7
- 229910052759 nickel Inorganic materials 0.000 description 7
- 238000003199 nucleic acid amplification method Methods 0.000 description 7
- 239000000047 product Substances 0.000 description 7
- 230000011664 signaling Effects 0.000 description 7
- 238000001262 western blot Methods 0.000 description 7
- 102000004190 Enzymes Human genes 0.000 description 6
- 108090000790 Enzymes Proteins 0.000 description 6
- 101001092197 Homo sapiens RNA binding protein fox-1 homolog 3 Proteins 0.000 description 6
- 241000699666 Mus <mouse, genus> Species 0.000 description 6
- 102100035530 RNA binding protein fox-1 homolog 3 Human genes 0.000 description 6
- 210000001638 cerebellum Anatomy 0.000 description 6
- 238000003776 cleavage reaction Methods 0.000 description 6
- 230000001186 cumulative effect Effects 0.000 description 6
- 229940088598 enzyme Drugs 0.000 description 6
- 210000001808 exosome Anatomy 0.000 description 6
- 239000013641 positive control Substances 0.000 description 6
- 230000008439 repair process Effects 0.000 description 6
- 230000007017 scission Effects 0.000 description 6
- 102000040650 (ribonucleotides)n+m Human genes 0.000 description 5
- 238000010453 CRISPR/Cas method Methods 0.000 description 5
- 108010047041 Complementarity Determining Regions Proteins 0.000 description 5
- 238000001712 DNA sequencing Methods 0.000 description 5
- 108050001049 Extracellular proteins Proteins 0.000 description 5
- 108010043121 Green Fluorescent Proteins Proteins 0.000 description 5
- 102000004144 Green Fluorescent Proteins Human genes 0.000 description 5
- 101000823051 Homo sapiens Amyloid-beta precursor protein Proteins 0.000 description 5
- 108010036933 Presenilin-1 Proteins 0.000 description 5
- 102000012412 Presenilin-1 Human genes 0.000 description 5
- 230000004075 alteration Effects 0.000 description 5
- 238000004587 chromatography analysis Methods 0.000 description 5
- 238000009826 distribution Methods 0.000 description 5
- 238000002474 experimental method Methods 0.000 description 5
- 239000005090 green fluorescent protein Substances 0.000 description 5
- 239000000543 intermediate Substances 0.000 description 5
- 210000001519 tissue Anatomy 0.000 description 5
- 102000000872 ATM Human genes 0.000 description 4
- 102100022204 DNA-dependent protein kinase catalytic subunit Human genes 0.000 description 4
- 101710157074 DNA-dependent protein kinase catalytic subunit Proteins 0.000 description 4
- 108010014303 DNA-directed DNA polymerase Proteins 0.000 description 4
- 102000016928 DNA-directed DNA polymerase Human genes 0.000 description 4
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 4
- 102000003960 Ligases Human genes 0.000 description 4
- 108090000364 Ligases Proteins 0.000 description 4
- 101710163270 Nuclease Proteins 0.000 description 4
- 108010068097 Rad51 Recombinase Proteins 0.000 description 4
- 102000002490 Rad51 Recombinase Human genes 0.000 description 4
- 238000007792 addition Methods 0.000 description 4
- 230000015572 biosynthetic process Effects 0.000 description 4
- 238000001514 detection method Methods 0.000 description 4
- 230000006870 function Effects 0.000 description 4
- 230000002068 genetic effect Effects 0.000 description 4
- 230000002401 inhibitory effect Effects 0.000 description 4
- 125000005647 linker group Chemical group 0.000 description 4
- 230000001537 neural effect Effects 0.000 description 4
- 239000013612 plasmid Substances 0.000 description 4
- 230000008569 process Effects 0.000 description 4
- 108091008146 restriction endonucleases Proteins 0.000 description 4
- PYWVYCXTNDRMGF-UHFFFAOYSA-N rhodamine B Chemical compound [Cl-].C=12C=CC(=[N+](CC)CC)C=C2OC2=CC(N(CC)CC)=CC=C2C=1C1=CC=CC=C1C(O)=O PYWVYCXTNDRMGF-UHFFFAOYSA-N 0.000 description 4
- 230000019491 signal transduction Effects 0.000 description 4
- 239000007787 solid Substances 0.000 description 4
- 230000008685 targeting Effects 0.000 description 4
- 230000014616 translation Effects 0.000 description 4
- 230000003442 weekly effect Effects 0.000 description 4
- 108010048112 Amyloidogenic Proteins Proteins 0.000 description 3
- 108091033409 CRISPR Proteins 0.000 description 3
- 101000777293 Homo sapiens Serine/threonine-protein kinase Chk1 Proteins 0.000 description 3
- 101000904868 Homo sapiens Transcriptional regulator ATRX Proteins 0.000 description 3
- 108010054477 Immunoglobulin Fab Fragments Proteins 0.000 description 3
- 102000001706 Immunoglobulin Fab Fragments Human genes 0.000 description 3
- 102100034343 Integrase Human genes 0.000 description 3
- KFZMGEQAYNKOFK-UHFFFAOYSA-N Isopropanol Chemical compound CC(C)O KFZMGEQAYNKOFK-UHFFFAOYSA-N 0.000 description 3
- OKKJLVBELUTLKV-UHFFFAOYSA-N Methanol Chemical compound OC OKKJLVBELUTLKV-UHFFFAOYSA-N 0.000 description 3
- 108091092724 Noncoding DNA Proteins 0.000 description 3
- 108091093037 Peptide nucleic acid Proteins 0.000 description 3
- 108010064218 Poly (ADP-Ribose) Polymerase-1 Proteins 0.000 description 3
- 102100023712 Poly [ADP-ribose] polymerase 1 Human genes 0.000 description 3
- 102100022033 Presenilin-1 Human genes 0.000 description 3
- 102000000574 RNA-Induced Silencing Complex Human genes 0.000 description 3
- 108010016790 RNA-Induced Silencing Complex Proteins 0.000 description 3
- 108700008625 Reporter Genes Proteins 0.000 description 3
- 108091081021 Sense strand Proteins 0.000 description 3
- 102100031081 Serine/threonine-protein kinase Chk1 Human genes 0.000 description 3
- 102100023931 Transcriptional regulator ATRX Human genes 0.000 description 3
- 238000004458 analytical method Methods 0.000 description 3
- 239000005557 antagonist Substances 0.000 description 3
- 239000002787 antisense oligonuctleotide Substances 0.000 description 3
- 210000003169 central nervous system Anatomy 0.000 description 3
- 125000000151 cysteine group Chemical class N[C@@H](CS)C(=O)* 0.000 description 3
- 230000001419 dependent effect Effects 0.000 description 3
- 230000029087 digestion Effects 0.000 description 3
- 239000003623 enhancer Substances 0.000 description 3
- 238000010348 incorporation Methods 0.000 description 3
- 238000009830 intercalation Methods 0.000 description 3
- 238000002595 magnetic resonance imaging Methods 0.000 description 3
- 238000004519 manufacturing process Methods 0.000 description 3
- 108091027963 non-coding RNA Proteins 0.000 description 3
- 102000042567 non-coding RNA Human genes 0.000 description 3
- 108010058731 nopaline synthase Proteins 0.000 description 3
- 125000002467 phosphate group Chemical group [H]OP(=O)(O[H])O[*] 0.000 description 3
- PTMHPRAIXMAOOB-UHFFFAOYSA-L phosphoramidate Chemical compound NP([O-])([O-])=O PTMHPRAIXMAOOB-UHFFFAOYSA-L 0.000 description 3
- 210000002442 prefrontal cortex Anatomy 0.000 description 3
- 230000002360 prefrontal effect Effects 0.000 description 3
- 238000002360 preparation method Methods 0.000 description 3
- 238000012800 visualization Methods 0.000 description 3
- 108091005957 yellow fluorescent proteins Proteins 0.000 description 3
- FBKFDSUZBVDZIX-FFGDOFBPSA-N (4s)-4-[[(2s)-2-[[(2s)-2-[[2-[[(2s)-2-[[(2s)-2-acetamido-3-phenylpropanoyl]amino]-4-methylsulfanylbutanoyl]amino]-2-methylpropanoyl]amino]-3-[4-(phosphonomethyl)phenyl]propanoyl]amino]-3-(6-chloro-1h-indol-3-yl)propanoyl]amino]-5-[[1-[[(2s)-1-amino-4-meth Chemical compound C([C@@H](C(=O)N[C@@H](CCSC)C(=O)NC(C)(C)C(=O)N[C@@H](CC=1C=CC(CP(O)(O)=O)=CC=1)C(=O)N[C@@H](CC=1C2=CC=C(Cl)C=C2NC=1)C(=O)N[C@@H](CCC(O)=O)C(=O)NC1(CC1)C(=O)N[C@@H](CC(C)C)C(N)=O)NC(C)=O)C1=CC=CC=C1 FBKFDSUZBVDZIX-FFGDOFBPSA-N 0.000 description 2
- FZWGECJQACGGTI-UHFFFAOYSA-N 2-amino-7-methyl-1,7-dihydro-6H-purin-6-one Chemical compound NC1=NC(O)=C2N(C)C=NC2=N1 FZWGECJQACGGTI-UHFFFAOYSA-N 0.000 description 2
- FWBHETKCLVMNFS-UHFFFAOYSA-N 4',6-Diamino-2-phenylindol Chemical compound C1=CC(C(=N)N)=CC=C1C1=CC2=CC=C(C(N)=N)C=C2N1 FWBHETKCLVMNFS-UHFFFAOYSA-N 0.000 description 2
- OIVLITBTBDPEFK-UHFFFAOYSA-N 5,6-dihydrouracil Chemical compound O=C1CCNC(=O)N1 OIVLITBTBDPEFK-UHFFFAOYSA-N 0.000 description 2
- LRFVTYWOQMYALW-UHFFFAOYSA-N 9H-xanthine Chemical compound O=C1NC(=O)NC2=C1NC=N2 LRFVTYWOQMYALW-UHFFFAOYSA-N 0.000 description 2
- 102000014156 AMP-Activated Protein Kinases Human genes 0.000 description 2
- 108010011376 AMP-Activated Protein Kinases Proteins 0.000 description 2
- 108010000700 Acetolactate synthase Proteins 0.000 description 2
- 102000002260 Alkaline Phosphatase Human genes 0.000 description 2
- 108020004774 Alkaline Phosphatase Proteins 0.000 description 2
- 102000009091 Amyloidogenic Proteins Human genes 0.000 description 2
- KAKZBPTYRLMSJV-UHFFFAOYSA-N Butadiene Chemical compound C=CC=C KAKZBPTYRLMSJV-UHFFFAOYSA-N 0.000 description 2
- KXDHJXZQYSOELW-UHFFFAOYSA-M Carbamate Chemical compound NC([O-])=O KXDHJXZQYSOELW-UHFFFAOYSA-M 0.000 description 2
- 108010035563 Chloramphenicol O-acetyltransferase Proteins 0.000 description 2
- HEDRZPFGACZZDS-UHFFFAOYSA-N Chloroform Chemical compound ClC(Cl)Cl HEDRZPFGACZZDS-UHFFFAOYSA-N 0.000 description 2
- 108010077544 Chromatin Proteins 0.000 description 2
- 102000008169 Co-Repressor Proteins Human genes 0.000 description 2
- 108010060434 Co-Repressor Proteins Proteins 0.000 description 2
- 108091035707 Consensus sequence Proteins 0.000 description 2
- 108010066133 D-octopine dehydrogenase Proteins 0.000 description 2
- 102000053602 DNA Human genes 0.000 description 2
- 102100027828 DNA repair protein XRCC4 Human genes 0.000 description 2
- 206010012289 Dementia Diseases 0.000 description 2
- 201000010374 Down Syndrome Diseases 0.000 description 2
- 102100031780 Endonuclease Human genes 0.000 description 2
- 108010042407 Endonucleases Proteins 0.000 description 2
- 101100264215 Gallus gallus XRCC6 gene Proteins 0.000 description 2
- 102000053187 Glucuronidase Human genes 0.000 description 2
- 108010060309 Glucuronidase Proteins 0.000 description 2
- 108090000246 Histone acetyltransferases Proteins 0.000 description 2
- 102000003893 Histone acetyltransferases Human genes 0.000 description 2
- 102000003964 Histone deacetylase Human genes 0.000 description 2
- 108090000353 Histone deacetylase Proteins 0.000 description 2
- 101000649315 Homo sapiens DNA repair protein XRCC4 Proteins 0.000 description 2
- 108010001336 Horseradish Peroxidase Proteins 0.000 description 2
- 102100023915 Insulin Human genes 0.000 description 2
- 101710203526 Integrase Proteins 0.000 description 2
- 238000012313 Kruskal-Wallis test Methods 0.000 description 2
- 108060001084 Luciferase Proteins 0.000 description 2
- 239000005089 Luciferase Substances 0.000 description 2
- 241000699670 Mus sp. Species 0.000 description 2
- YJDAOHJWLUNFLX-UHFFFAOYSA-N NU 1025 Chemical compound C1=CC=C2C(=O)NC(C)=NC2=C1O YJDAOHJWLUNFLX-UHFFFAOYSA-N 0.000 description 2
- 108091093105 Nuclear DNA Proteins 0.000 description 2
- ISWSIDIOOBJBQZ-UHFFFAOYSA-N Phenol Chemical compound OC1=CC=CC=C1 ISWSIDIOOBJBQZ-UHFFFAOYSA-N 0.000 description 2
- 102000001253 Protein Kinase Human genes 0.000 description 2
- 108020005067 RNA Splice Sites Proteins 0.000 description 2
- 108010041191 Sirtuin 1 Proteins 0.000 description 2
- 102000013530 TOR Serine-Threonine Kinases Human genes 0.000 description 2
- 108010065917 TOR Serine-Threonine Kinases Proteins 0.000 description 2
- 108091046869 Telomeric non-coding RNA Proteins 0.000 description 2
- 102000040945 Transcription factor Human genes 0.000 description 2
- 108091023040 Transcription factor Proteins 0.000 description 2
- ISAKRJDGNUQOIC-UHFFFAOYSA-N Uracil Chemical compound O=C1C=CNC(=O)N1 ISAKRJDGNUQOIC-UHFFFAOYSA-N 0.000 description 2
- 102100036976 X-ray repair cross-complementing protein 6 Human genes 0.000 description 2
- MWPLVEDNUUSJAV-UHFFFAOYSA-N anthracene Chemical compound C1=CC=CC2=CC3=CC=CC=C3C=C21 MWPLVEDNUUSJAV-UHFFFAOYSA-N 0.000 description 2
- 210000001130 astrocyte Anatomy 0.000 description 2
- ZYGHJZDHTFUPRJ-UHFFFAOYSA-N benzo-alpha-pyrone Natural products C1=CC=C2OC(=O)C=CC2=C1 ZYGHJZDHTFUPRJ-UHFFFAOYSA-N 0.000 description 2
- 230000008499 blood brain barrier function Effects 0.000 description 2
- 210000001218 blood-brain barrier Anatomy 0.000 description 2
- RYYVLZVUVIJVGH-UHFFFAOYSA-N caffeine Chemical group CN1C(=O)N(C)C(=O)C2=C1N=CN2C RYYVLZVUVIJVGH-UHFFFAOYSA-N 0.000 description 2
- 230000003197 catalytic effect Effects 0.000 description 2
- 238000005119 centrifugation Methods 0.000 description 2
- 239000013522 chelant Substances 0.000 description 2
- HWGQMRYQVZSGDQ-HZPDHXFCSA-N chembl3137320 Chemical compound CN1N=CN=C1[C@H]([C@H](N1)C=2C=CC(F)=CC=2)C2=NNC(=O)C3=C2C1=CC(F)=C3 HWGQMRYQVZSGDQ-HZPDHXFCSA-N 0.000 description 2
- 210000003483 chromatin Anatomy 0.000 description 2
- 230000003081 coactivator Effects 0.000 description 2
- VPUGDVKSAQVFFS-UHFFFAOYSA-N coronene Chemical compound C1=C(C2=C34)C=CC3=CC=C(C=C3)C4=C4C3=CC=C(C=C3)C4=C2C3=C1 VPUGDVKSAQVFFS-UHFFFAOYSA-N 0.000 description 2
- 238000012937 correction Methods 0.000 description 2
- 235000001671 coumarin Nutrition 0.000 description 2
- 108010082025 cyan fluorescent protein Proteins 0.000 description 2
- 235000018417 cysteine Nutrition 0.000 description 2
- OPTASPLRGRRNAP-UHFFFAOYSA-N cytosine Chemical compound NC=1C=CNC(=O)N=1 OPTASPLRGRRNAP-UHFFFAOYSA-N 0.000 description 2
- 210000004443 dendritic cell Anatomy 0.000 description 2
- 238000007847 digital PCR Methods 0.000 description 2
- 239000000539 dimer Substances 0.000 description 2
- 238000011304 droplet digital PCR Methods 0.000 description 2
- 238000001962 electrophoresis Methods 0.000 description 2
- 238000005516 engineering process Methods 0.000 description 2
- MHMNJMPURVTYEJ-UHFFFAOYSA-N fluorescein-5-isothiocyanate Chemical compound O1C(=O)C2=CC(N=C=S)=CC=C2C21C1=CC=C(O)C=C1OC1=CC(O)=CC=C21 MHMNJMPURVTYEJ-UHFFFAOYSA-N 0.000 description 2
- 238000001943 fluorescence-activated cell sorting Methods 0.000 description 2
- UYTPUPDQBNUYGX-UHFFFAOYSA-N guanine Chemical compound O=C1NC(N)=NC2=C1N=CN2 UYTPUPDQBNUYGX-UHFFFAOYSA-N 0.000 description 2
- 102000046783 human APP Human genes 0.000 description 2
- FDGQSTZJBFJUBT-UHFFFAOYSA-N hypoxanthine Chemical compound O=C1NC=NC2=C1NC=N2 FDGQSTZJBFJUBT-UHFFFAOYSA-N 0.000 description 2
- 238000010166 immunofluorescence Methods 0.000 description 2
- 238000001427 incoherent neutron scattering Methods 0.000 description 2
- 238000005304 joining Methods 0.000 description 2
- 101150085005 ku70 gene Proteins 0.000 description 2
- 239000003550 marker Substances 0.000 description 2
- 239000000463 material Substances 0.000 description 2
- 230000001404 mediated effect Effects 0.000 description 2
- 230000003340 mental effect Effects 0.000 description 2
- 210000000274 microglia Anatomy 0.000 description 2
- 230000033607 mismatch repair Effects 0.000 description 2
- 125000004573 morpholin-4-yl group Chemical group N1(CCOCC1)* 0.000 description 2
- 238000002610 neuroimaging Methods 0.000 description 2
- 230000000926 neurological effect Effects 0.000 description 2
- 230000007171 neuropathology Effects 0.000 description 2
- 230000020520 nucleotide-excision repair Effects 0.000 description 2
- 230000001717 pathogenic effect Effects 0.000 description 2
- 108091007428 primary miRNA Proteins 0.000 description 2
- 238000012545 processing Methods 0.000 description 2
- 108060006633 protein kinase Proteins 0.000 description 2
- BBEAQIROQSPTKN-UHFFFAOYSA-N pyrene Chemical compound C1=CC=C2C=CC3=CC=CC4=CC=C1C2=C43 BBEAQIROQSPTKN-UHFFFAOYSA-N 0.000 description 2
- 238000011002 quantification Methods 0.000 description 2
- 102000005962 receptors Human genes 0.000 description 2
- 108020003175 receptors Proteins 0.000 description 2
- 230000006798 recombination Effects 0.000 description 2
- 238000005215 recombination Methods 0.000 description 2
- 108010054624 red fluorescent protein Proteins 0.000 description 2
- 230000002829 reductive effect Effects 0.000 description 2
- 238000010839 reverse transcription Methods 0.000 description 2
- 125000002652 ribonucleotide group Chemical group 0.000 description 2
- HMABYWSNWIZPAG-UHFFFAOYSA-N rucaparib Chemical compound C1=CC(CNC)=CC=C1C(N1)=C2CCNC(=O)C3=C2C1=CC(F)=C3 HMABYWSNWIZPAG-UHFFFAOYSA-N 0.000 description 2
- 229950004707 rucaparib Drugs 0.000 description 2
- 230000003584 silencer Effects 0.000 description 2
- 210000001324 spliceosome Anatomy 0.000 description 2
- PVYJZLYGTZKPJE-UHFFFAOYSA-N streptonigrin Chemical compound C=1C=C2C(=O)C(OC)=C(N)C(=O)C2=NC=1C(C=1N)=NC(C(O)=O)=C(C)C=1C1=CC=C(OC)C(OC)=C1O PVYJZLYGTZKPJE-UHFFFAOYSA-N 0.000 description 2
- 239000000126 substance Substances 0.000 description 2
- 238000006467 substitution reaction Methods 0.000 description 2
- 150000008163 sugars Chemical class 0.000 description 2
- MPLHNVLQVRSVEE-UHFFFAOYSA-N texas red Chemical compound [O-]S(=O)(=O)C1=CC(S(Cl)(=O)=O)=CC=C1C(C1=CC=2CCCN3CCCC(C=23)=C1O1)=C2C1=C(CCC1)C3=[N+]1CCCC3=C2 MPLHNVLQVRSVEE-UHFFFAOYSA-N 0.000 description 2
- RYYWUUFWQRZTIU-UHFFFAOYSA-K thiophosphate Chemical compound [O-]P([O-])([O-])=S RYYWUUFWQRZTIU-UHFFFAOYSA-K 0.000 description 2
- RWQNBRDOKXIBIV-UHFFFAOYSA-N thymine Chemical compound CC1=CNC(=O)NC1=O RWQNBRDOKXIBIV-UHFFFAOYSA-N 0.000 description 2
- 238000010361 transduction Methods 0.000 description 2
- 230000026683 transduction Effects 0.000 description 2
- 238000013519 translation Methods 0.000 description 2
- 230000005945 translocation Effects 0.000 description 2
- DENYZIUJOTUUNY-MRXNPFEDSA-N (2R)-14-fluoro-2-methyl-6,9,10,19-tetrazapentacyclo[14.2.1.02,6.08,18.012,17]nonadeca-1(18),8,12(17),13,15-pentaen-11-one Chemical compound FC=1C=C2C=3C=4C(CN5[C@@](C4NC3C1)(CCC5)C)=NNC2=O DENYZIUJOTUUNY-MRXNPFEDSA-N 0.000 description 1
- QGKMIGUHVLGJBR-UHFFFAOYSA-M (4z)-1-(3-methylbutyl)-4-[[1-(3-methylbutyl)quinolin-1-ium-4-yl]methylidene]quinoline;iodide Chemical compound [I-].C12=CC=CC=C2N(CCC(C)C)C=CC1=CC1=CC=[N+](CCC(C)C)C2=CC=CC=C12 QGKMIGUHVLGJBR-UHFFFAOYSA-M 0.000 description 1
- MOWXJLUYGFNTAL-DEOSSOPVSA-N (s)-[2-chloro-4-fluoro-5-(7-morpholin-4-ylquinazolin-4-yl)phenyl]-(6-methoxypyridazin-3-yl)methanol Chemical compound N1=NC(OC)=CC=C1[C@@H](O)C1=CC(C=2C3=CC=C(C=C3N=CN=2)N2CCOCC2)=C(F)C=C1Cl MOWXJLUYGFNTAL-DEOSSOPVSA-N 0.000 description 1
- CTLOSZHDGZLOQE-UHFFFAOYSA-N 14-methoxy-9-[(4-methylpiperazin-1-yl)methyl]-9,19-diazapentacyclo[10.7.0.02,6.07,11.013,18]nonadeca-1(12),2(6),7(11),13(18),14,16-hexaene-8,10-dione Chemical compound O=C1C2=C3C=4C(OC)=CC=CC=4NC3=C3CCCC3=C2C(=O)N1CN1CCN(C)CC1 CTLOSZHDGZLOQE-UHFFFAOYSA-N 0.000 description 1
- YMHOBZXQZVXHBM-UHFFFAOYSA-N 2,5-dimethoxy-4-bromophenethylamine Chemical compound COC1=CC(CCN)=C(OC)C=C1Br YMHOBZXQZVXHBM-UHFFFAOYSA-N 0.000 description 1
- OBYNJKLOYWCXEP-UHFFFAOYSA-N 2-[3-(dimethylamino)-6-dimethylazaniumylidenexanthen-9-yl]-4-isothiocyanatobenzoate Chemical compound C=12C=CC(=[N+](C)C)C=C2OC2=CC(N(C)C)=CC=C2C=1C1=CC(N=C=S)=CC=C1C([O-])=O OBYNJKLOYWCXEP-UHFFFAOYSA-N 0.000 description 1
- GOJUJUVQIVIZAV-UHFFFAOYSA-N 2-amino-4,6-dichloropyrimidine-5-carbaldehyde Chemical group NC1=NC(Cl)=C(C=O)C(Cl)=N1 GOJUJUVQIVIZAV-UHFFFAOYSA-N 0.000 description 1
- ASJSAQIRZKANQN-CRCLSJGQSA-N 2-deoxy-D-ribose Chemical compound OC[C@@H](O)[C@@H](O)CC=O ASJSAQIRZKANQN-CRCLSJGQSA-N 0.000 description 1
- KYGSXEYUWRFVNY-UHFFFAOYSA-N 2-pyran-2-ylidenepropanedinitrile Chemical class N#CC(C#N)=C1OC=CC=C1 KYGSXEYUWRFVNY-UHFFFAOYSA-N 0.000 description 1
- OALHHIHQOFIMEF-UHFFFAOYSA-N 3',6'-dihydroxy-2',4',5',7'-tetraiodo-3h-spiro[2-benzofuran-1,9'-xanthene]-3-one Chemical compound O1C(=O)C2=CC=CC=C2C21C1=CC(I)=C(O)C(I)=C1OC1=C(I)C(O)=C(I)C=C21 OALHHIHQOFIMEF-UHFFFAOYSA-N 0.000 description 1
- GSCPDZHWVNUUFI-UHFFFAOYSA-N 3-aminobenzamide Chemical compound NC(=O)C1=CC=CC(N)=C1 GSCPDZHWVNUUFI-UHFFFAOYSA-N 0.000 description 1
- FWMNVWWHGCHHJJ-SKKKGAJSSA-N 4-amino-1-[(2r)-6-amino-2-[[(2r)-2-[[(2r)-2-[[(2r)-2-amino-3-phenylpropanoyl]amino]-3-phenylpropanoyl]amino]-4-methylpentanoyl]amino]hexanoyl]piperidine-4-carboxylic acid Chemical compound C([C@H](C(=O)N[C@H](CC(C)C)C(=O)N[C@H](CCCCN)C(=O)N1CCC(N)(CC1)C(O)=O)NC(=O)[C@H](N)CC=1C=CC=CC=1)C1=CC=CC=C1 FWMNVWWHGCHHJJ-SKKKGAJSSA-N 0.000 description 1
- 229960000549 4-dimethylaminophenol Drugs 0.000 description 1
- VHYFNPMBLIVWCW-UHFFFAOYSA-N 4-dimethylaminopyridine Substances CN(C)C1=CC=NC=C1 VHYFNPMBLIVWCW-UHFFFAOYSA-N 0.000 description 1
- MDOJTZQKHMAPBK-UHFFFAOYSA-N 4-iodo-3-nitrobenzamide Chemical compound NC(=O)C1=CC=C(I)C([N+]([O-])=O)=C1 MDOJTZQKHMAPBK-UHFFFAOYSA-N 0.000 description 1
- UPJKSWLLCONYMW-UHFFFAOYSA-N 5'-Adenosine monophosphate Natural products COc1cc(O)c(C(=O)C)c(OC2OC(COC3OC(C)C(O)C(O)C3O)C(O)C(O)C2O)c1 UPJKSWLLCONYMW-UHFFFAOYSA-N 0.000 description 1
- NEEVCWPRIZJJRJ-LWRDCAMISA-N 5-(benzylideneamino)-6-[(e)-benzylideneamino]-2-sulfanylidene-1h-pyrimidin-4-one Chemical compound C=1C=CC=CC=1C=NC=1C(=O)NC(=S)NC=1\N=C\C1=CC=CC=C1 NEEVCWPRIZJJRJ-LWRDCAMISA-N 0.000 description 1
- BLQMCTXZEMGOJM-UHFFFAOYSA-N 5-carboxycytosine Chemical compound NC=1NC(=O)N=CC=1C(O)=O BLQMCTXZEMGOJM-UHFFFAOYSA-N 0.000 description 1
- NJYVEMPWNAYQQN-UHFFFAOYSA-N 5-carboxyfluorescein Chemical compound C12=CC=C(O)C=C2OC2=CC(O)=CC=C2C21OC(=O)C1=CC(C(=O)O)=CC=C21 NJYVEMPWNAYQQN-UHFFFAOYSA-N 0.000 description 1
- YMZMTOFQCVHHFB-UHFFFAOYSA-N 5-carboxytetramethylrhodamine Chemical compound C=12C=CC(N(C)C)=CC2=[O+]C2=CC(N(C)C)=CC=C2C=1C1=CC=C(C(O)=O)C=C1C([O-])=O YMZMTOFQCVHHFB-UHFFFAOYSA-N 0.000 description 1
- LRSASMSXMSNRBT-UHFFFAOYSA-N 5-methylcytosine Chemical compound CC1=CNC(=O)N=C1N LRSASMSXMSNRBT-UHFFFAOYSA-N 0.000 description 1
- BZTDTCNHAFUJOG-UHFFFAOYSA-N 6-carboxyfluorescein Chemical compound C12=CC=C(O)C=C2OC2=CC(O)=CC=C2C11OC(=O)C2=CC=C(C(=O)O)C=C21 BZTDTCNHAFUJOG-UHFFFAOYSA-N 0.000 description 1
- CJIJXIFQYOPWTF-UHFFFAOYSA-N 7-hydroxycoumarin Natural products O1C(=O)C=CC2=CC(O)=CC=C21 CJIJXIFQYOPWTF-UHFFFAOYSA-N 0.000 description 1
- PBCZSGKMGDDXIJ-HQCWYSJUSA-N 7-hydroxystaurosporine Chemical compound N([C@H](O)C1=C2C3=CC=CC=C3N3C2=C24)C(=O)C1=C2C1=CC=CC=C1N4[C@H]1C[C@@H](NC)[C@@H](OC)[C@]3(C)O1 PBCZSGKMGDDXIJ-HQCWYSJUSA-N 0.000 description 1
- PBCZSGKMGDDXIJ-UHFFFAOYSA-N 7beta-hydroxystaurosporine Natural products C12=C3N4C5=CC=CC=C5C3=C3C(O)NC(=O)C3=C2C2=CC=CC=C2N1C1CC(NC)C(OC)C4(C)O1 PBCZSGKMGDDXIJ-UHFFFAOYSA-N 0.000 description 1
- GJCOSYZMQJWQCA-UHFFFAOYSA-N 9H-xanthene Chemical compound C1=CC=C2CC3=CC=CC=C3OC2=C1 GJCOSYZMQJWQCA-UHFFFAOYSA-N 0.000 description 1
- 241000604451 Acidaminococcus Species 0.000 description 1
- GDALETGZDYOOGB-UHFFFAOYSA-N Acridone Natural products C1=C(O)C=C2N(C)C3=CC=CC=C3C(=O)C2=C1O GDALETGZDYOOGB-UHFFFAOYSA-N 0.000 description 1
- GFFGJBXGBJISGV-UHFFFAOYSA-N Adenine Chemical compound NC1=NC=NC2=C1N=CN2 GFFGJBXGBJISGV-UHFFFAOYSA-N 0.000 description 1
- 229930024421 Adenine Natural products 0.000 description 1
- 239000012118 Alexa Fluor 750 Substances 0.000 description 1
- 241001147780 Alicyclobacillus Species 0.000 description 1
- 238000010173 Alzheimer-disease mouse model Methods 0.000 description 1
- USFZMSVCRYTOJT-UHFFFAOYSA-N Ammonium acetate Chemical compound N.CC(O)=O USFZMSVCRYTOJT-UHFFFAOYSA-N 0.000 description 1
- 239000005695 Ammonium acetate Substances 0.000 description 1
- 102000002659 Amyloid Precursor Protein Secretases Human genes 0.000 description 1
- 102000008682 Argonaute Proteins Human genes 0.000 description 1
- 108010088141 Argonaute Proteins Proteins 0.000 description 1
- 241000589941 Azospirillum Species 0.000 description 1
- 108091005950 Azurite Proteins 0.000 description 1
- 102000036365 BRCA1 Human genes 0.000 description 1
- 108700020463 BRCA1 Proteins 0.000 description 1
- 101150072950 BRCA1 gene Proteins 0.000 description 1
- 102000052609 BRCA2 Human genes 0.000 description 1
- 108700020462 BRCA2 Proteins 0.000 description 1
- 241000193830 Bacillus <bacterium> Species 0.000 description 1
- 241000605059 Bacteroidetes Species 0.000 description 1
- 101150008921 Brca2 gene Proteins 0.000 description 1
- 108010040467 CRISPR-Associated Proteins Proteins 0.000 description 1
- 101150018129 CSF2 gene Proteins 0.000 description 1
- 101150069031 CSN2 gene Proteins 0.000 description 1
- 102000000905 Cadherin Human genes 0.000 description 1
- 108050007957 Cadherin Proteins 0.000 description 1
- 241000589876 Campylobacter Species 0.000 description 1
- 102100025570 Cancer/testis antigen 1 Human genes 0.000 description 1
- 241000206594 Carnobacterium Species 0.000 description 1
- 102000016289 Cell Adhesion Molecules Human genes 0.000 description 1
- 108010067225 Cell Adhesion Molecules Proteins 0.000 description 1
- 108091005944 Cerulean Proteins 0.000 description 1
- 241000195585 Chlamydomonas Species 0.000 description 1
- 108010009685 Cholinergic Receptors Proteins 0.000 description 1
- 108091005960 Citrine Proteins 0.000 description 1
- 241000193403 Clostridium Species 0.000 description 1
- UDMBCSSLTHHNCD-UHFFFAOYSA-N Coenzym Q(11) Natural products C1=NC=2C(N)=NC=NC=2N1C1OC(COP(O)(O)=O)C(O)C1O UDMBCSSLTHHNCD-UHFFFAOYSA-N 0.000 description 1
- 108020004635 Complementary DNA Proteins 0.000 description 1
- 101150074775 Csf1 gene Proteins 0.000 description 1
- 108091005943 CyPet Proteins 0.000 description 1
- 102000004127 Cytokines Human genes 0.000 description 1
- 108090000695 Cytokines Proteins 0.000 description 1
- HMFHBZSHGGEWLO-SOOFDHNKSA-N D-ribofuranose Chemical compound OC[C@H]1OC(O)[C@H](O)[C@@H]1O HMFHBZSHGGEWLO-SOOFDHNKSA-N 0.000 description 1
- 238000000116 DAPI staining Methods 0.000 description 1
- 230000005778 DNA damage Effects 0.000 description 1
- 231100000277 DNA damage Toxicity 0.000 description 1
- 102100035186 DNA excision repair protein ERCC-1 Human genes 0.000 description 1
- 102100029765 DNA polymerase lambda Human genes 0.000 description 1
- 101710177421 DNA polymerase lambda Proteins 0.000 description 1
- 108010061914 DNA polymerase mu Proteins 0.000 description 1
- 102100029764 DNA-directed DNA/RNA polymerase mu Human genes 0.000 description 1
- 102000004163 DNA-directed RNA polymerases Human genes 0.000 description 1
- 108090000626 DNA-directed RNA polymerases Proteins 0.000 description 1
- 102000009058 Death Domain Receptors Human genes 0.000 description 1
- 108010049207 Death Domain Receptors Proteins 0.000 description 1
- 241000936939 Desulfonatronum Species 0.000 description 1
- 241000605716 Desulfovibrio Species 0.000 description 1
- 241000205236 Desulfurococcus Species 0.000 description 1
- BWGNESOTFCXPMA-UHFFFAOYSA-N Dihydrogen disulfide Chemical compound SS BWGNESOTFCXPMA-UHFFFAOYSA-N 0.000 description 1
- 102100033996 Double-strand break repair protein MRE11 Human genes 0.000 description 1
- 108091005941 EBFP Proteins 0.000 description 1
- 108091005947 EBFP2 Proteins 0.000 description 1
- 108091005942 ECFP Proteins 0.000 description 1
- 239000005977 Ethylene Substances 0.000 description 1
- 241000186394 Eubacterium Species 0.000 description 1
- 241000589565 Flavobacterium Species 0.000 description 1
- 241000589601 Francisella Species 0.000 description 1
- 102000003688 G-Protein-Coupled Receptors Human genes 0.000 description 1
- 108090000045 G-Protein-Coupled Receptors Proteins 0.000 description 1
- 101150106478 GPS1 gene Proteins 0.000 description 1
- 241000032681 Gluconacetobacter Species 0.000 description 1
- 241001430278 Helcococcus Species 0.000 description 1
- 108010036115 Histone Methyltransferases Proteins 0.000 description 1
- 241000282412 Homo Species 0.000 description 1
- 101000856237 Homo sapiens Cancer/testis antigen 1 Proteins 0.000 description 1
- 101000876529 Homo sapiens DNA excision repair protein ERCC-1 Proteins 0.000 description 1
- 101000591400 Homo sapiens Double-strand break repair protein MRE11 Proteins 0.000 description 1
- 101000611202 Homo sapiens Peptidyl-prolyl cis-trans isomerase B Proteins 0.000 description 1
- 101001123331 Homo sapiens Peroxisome proliferator-activated receptor gamma coactivator 1-alpha Proteins 0.000 description 1
- UGQMRVRMYYASKQ-UHFFFAOYSA-N Hypoxanthine nucleoside Natural products OC1C(O)C(CO)OC1N1C(NC=NC2=O)=C2N=C1 UGQMRVRMYYASKQ-UHFFFAOYSA-N 0.000 description 1
- DGAQECJNVWCQMB-PUAWFVPOSA-M Ilexoside XXIX Chemical compound C[C@@H]1CC[C@@]2(CC[C@@]3(C(=CC[C@H]4[C@]3(CC[C@@H]5[C@@]4(CC[C@@H](C5(C)C)OS(=O)(=O)[O-])C)C)[C@@H]2[C@]1(C)O)C)C(=O)O[C@H]6[C@@H]([C@H]([C@@H]([C@H](O6)CO)O)O)O.[Na+] DGAQECJNVWCQMB-PUAWFVPOSA-M 0.000 description 1
- 102000012745 Immunoglobulin Subunits Human genes 0.000 description 1
- 108010079585 Immunoglobulin Subunits Proteins 0.000 description 1
- 108010067060 Immunoglobulin Variable Region Proteins 0.000 description 1
- 102000017727 Immunoglobulin Variable Region Human genes 0.000 description 1
- LPHGQDQBBGAPDZ-UHFFFAOYSA-N Isocaffeine Natural products CN1C(=O)N(C)C(=O)C2=C1N(C)C=N2 LPHGQDQBBGAPDZ-UHFFFAOYSA-N 0.000 description 1
- 238000001276 Kolmogorov–Smirnov test Methods 0.000 description 1
- 241001112693 Lachnospiraceae Species 0.000 description 1
- 241000186660 Lactobacillus Species 0.000 description 1
- 241000589248 Legionella Species 0.000 description 1
- 208000007764 Legionnaires' Disease Diseases 0.000 description 1
- 241001453171 Leptotrichia Species 0.000 description 1
- 241000186781 Listeria Species 0.000 description 1
- 241000589323 Methylobacterium Species 0.000 description 1
- YBHQCJILTOVLHD-YVMONPNESA-N Mirin Chemical compound S1C(N)=NC(=O)\C1=C\C1=CC=C(O)C=C1 YBHQCJILTOVLHD-YVMONPNESA-N 0.000 description 1
- 241001529936 Murinae Species 0.000 description 1
- 101100219625 Mus musculus Casd1 gene Proteins 0.000 description 1
- 241000204031 Mycoplasma Species 0.000 description 1
- 102100031455 NAD-dependent protein deacetylase sirtuin-1 Human genes 0.000 description 1
- 241000204974 Natronobacterium Species 0.000 description 1
- 241000588653 Neisseria Species 0.000 description 1
- 101100385413 Neurospora crassa (strain ATCC 24698 / 74-OR23-1A / CBS 708.71 / DSM 1257 / FGSC 987) csm-3 gene Proteins 0.000 description 1
- 241000135938 Nitratifractor Species 0.000 description 1
- 241000936936 Opitutaceae Species 0.000 description 1
- 229910019142 PO4 Inorganic materials 0.000 description 1
- 108010016731 PPAR gamma Proteins 0.000 description 1
- 241000740708 Paludibacter Species 0.000 description 1
- 108090000526 Papain Proteins 0.000 description 1
- 241001386753 Parvibaculum Species 0.000 description 1
- 102000057297 Pepsin A Human genes 0.000 description 1
- 108090000284 Pepsin A Proteins 0.000 description 1
- 102100040283 Peptidyl-prolyl cis-trans isomerase B Human genes 0.000 description 1
- 102100038825 Peroxisome proliferator-activated receptor gamma Human genes 0.000 description 1
- 102100028960 Peroxisome proliferator-activated receptor gamma coactivator 1-alpha Human genes 0.000 description 1
- 108010004729 Phycoerythrin Proteins 0.000 description 1
- 241000605894 Porphyromonas Species 0.000 description 1
- ZLMJMSJWJFRBEC-UHFFFAOYSA-N Potassium Chemical compound [K] ZLMJMSJWJFRBEC-UHFFFAOYSA-N 0.000 description 1
- 241000605861 Prevotella Species 0.000 description 1
- 239000004365 Protease Substances 0.000 description 1
- 108700040121 Protein Methyltransferases Proteins 0.000 description 1
- 102000055027 Protein Methyltransferases Human genes 0.000 description 1
- 241001323319 Psen Species 0.000 description 1
- 241000205160 Pyrococcus Species 0.000 description 1
- NRCMAYZCPIVABH-UHFFFAOYSA-N Quinacridone Chemical compound N1C2=CC=CC=C2C(=O)C2=C1C=C1C(=O)C3=CC=CC=C3NC1=C2 NRCMAYZCPIVABH-UHFFFAOYSA-N 0.000 description 1
- 108020004518 RNA Probes Proteins 0.000 description 1
- 239000003391 RNA probe Substances 0.000 description 1
- 108010092799 RNA-directed DNA polymerase Proteins 0.000 description 1
- 101100047461 Rattus norvegicus Trpm8 gene Proteins 0.000 description 1
- 241000191025 Rhodobacter Species 0.000 description 1
- 108091028664 Ribonucleotide Proteins 0.000 description 1
- PYMYPHUHKUWMLA-LMVFSUKVSA-N Ribose Natural products OC[C@@H](O)[C@@H](O)[C@@H](O)C=O PYMYPHUHKUWMLA-LMVFSUKVSA-N 0.000 description 1
- 241000605947 Roseburia Species 0.000 description 1
- CGNLCCVKSWNSDG-UHFFFAOYSA-N SYBR Green I Chemical compound CN(C)CCCN(CCC)C1=CC(C=C2N(C3=CC=CC=C3S2)C)=C2C=CC=CC2=[N+]1C1=CC=CC=C1 CGNLCCVKSWNSDG-UHFFFAOYSA-N 0.000 description 1
- 241000235070 Saccharomyces Species 0.000 description 1
- 102000000344 Sirtuin 1 Human genes 0.000 description 1
- 241000949716 Sphaerochaeta Species 0.000 description 1
- 241000191940 Staphylococcus Species 0.000 description 1
- PJANXHGTPQOBST-VAWYXSNFSA-N Stilbene Natural products C=1C=CC=CC=1/C=C/C1=CC=CC=C1 PJANXHGTPQOBST-VAWYXSNFSA-N 0.000 description 1
- 241000194017 Streptococcus Species 0.000 description 1
- 108091008874 T cell receptors Proteins 0.000 description 1
- 102000016266 T-Cell Antigen Receptors Human genes 0.000 description 1
- 241000589596 Thermus Species 0.000 description 1
- 108020004566 Transfer RNA Proteins 0.000 description 1
- 239000007984 Tris EDTA buffer Substances 0.000 description 1
- 239000007983 Tris buffer Substances 0.000 description 1
- 241000670722 Tuberibacillus Species 0.000 description 1
- 241000545067 Venus Species 0.000 description 1
- 102000034337 acetylcholine receptors Human genes 0.000 description 1
- DPKHZNPWBDQZCN-UHFFFAOYSA-N acridine orange free base Chemical compound C1=CC(N(C)C)=CC2=NC3=CC(N(C)C)=CC=C3C=C21 DPKHZNPWBDQZCN-UHFFFAOYSA-N 0.000 description 1
- FZEYVTFCMJSGMP-UHFFFAOYSA-N acridone Chemical compound C1=CC=C2C(=O)C3=CC=CC=C3NC2=C1 FZEYVTFCMJSGMP-UHFFFAOYSA-N 0.000 description 1
- 230000009471 action Effects 0.000 description 1
- 239000012190 activator Substances 0.000 description 1
- 229960000643 adenine Drugs 0.000 description 1
- UDMBCSSLTHHNCD-KQYNXXCUSA-N adenosine 5'-monophosphate Chemical compound C1=NC=2C(N)=NC=NC=2N1[C@@H]1O[C@H](COP(O)(O)=O)[C@@H](O)[C@H]1O UDMBCSSLTHHNCD-KQYNXXCUSA-N 0.000 description 1
- HMFHBZSHGGEWLO-UHFFFAOYSA-N alpha-D-Furanose-Ribose Natural products OCC1OC(O)C(O)C1O HMFHBZSHGGEWLO-UHFFFAOYSA-N 0.000 description 1
- 102000038380 alpha-secretases Human genes 0.000 description 1
- 108091007736 alpha-secretases Proteins 0.000 description 1
- 150000001408 amides Chemical class 0.000 description 1
- 235000019257 ammonium acetate Nutrition 0.000 description 1
- 229940043376 ammonium acetate Drugs 0.000 description 1
- 230000003942 amyloidogenic effect Effects 0.000 description 1
- 238000009175 antibody therapy Methods 0.000 description 1
- 150000004982 aromatic amines Chemical class 0.000 description 1
- 238000003491 array Methods 0.000 description 1
- 230000008901 benefit Effects 0.000 description 1
- DZBUGLKDJFMEHC-UHFFFAOYSA-N benzoquinolinylidene Natural products C1=CC=CC2=CC3=CC=CC=C3N=C21 DZBUGLKDJFMEHC-UHFFFAOYSA-N 0.000 description 1
- HMFHBZSHGGEWLO-TXICZTDVSA-N beta-D-ribose Chemical group OC[C@H]1O[C@@H](O)[C@H](O)[C@@H]1O HMFHBZSHGGEWLO-TXICZTDVSA-N 0.000 description 1
- 102000005936 beta-Galactosidase Human genes 0.000 description 1
- 108010005774 beta-Galactosidase Proteins 0.000 description 1
- 108091007737 beta-secretases Proteins 0.000 description 1
- 125000002619 bicyclic group Chemical class 0.000 description 1
- 230000003115 biocidal effect Effects 0.000 description 1
- 230000004071 biological effect Effects 0.000 description 1
- 210000001772 blood platelet Anatomy 0.000 description 1
- 210000004899 c-terminal region Anatomy 0.000 description 1
- AIYUHDOJVYHVIT-UHFFFAOYSA-M caesium chloride Chemical compound [Cl-].[Cs+] AIYUHDOJVYHVIT-UHFFFAOYSA-M 0.000 description 1
- 229960001948 caffeine Drugs 0.000 description 1
- VJEONQKOZGKCAK-UHFFFAOYSA-N caffeine Natural products CN1C(=O)N(C)C(=O)C2=C1C=CN2C VJEONQKOZGKCAK-UHFFFAOYSA-N 0.000 description 1
- 125000000609 carbazolyl group Chemical class C1(=CC=CC=2C3=CC=CC=C3NC12)* 0.000 description 1
- 101150055766 cat gene Proteins 0.000 description 1
- 230000015556 catabolic process Effects 0.000 description 1
- 239000013592 cell lysate Substances 0.000 description 1
- 230000006037 cell lysis Effects 0.000 description 1
- 210000000170 cell membrane Anatomy 0.000 description 1
- 238000012412 chemical coupling Methods 0.000 description 1
- 230000001684 chronic effect Effects 0.000 description 1
- 239000011035 citrine Substances 0.000 description 1
- 230000004186 co-expression Effects 0.000 description 1
- 230000008045 co-localization Effects 0.000 description 1
- 230000003920 cognitive function Effects 0.000 description 1
- 238000004737 colorimetric analysis Methods 0.000 description 1
- 238000003271 compound fluorescence assay Methods 0.000 description 1
- 238000002591 computed tomography Methods 0.000 description 1
- 101150055601 cops2 gene Proteins 0.000 description 1
- 210000005257 cortical tissue Anatomy 0.000 description 1
- 150000004775 coumarins Chemical class 0.000 description 1
- 150000001907 coumarones Chemical class 0.000 description 1
- 238000004132 cross linking Methods 0.000 description 1
- 125000000753 cycloalkyl group Chemical group 0.000 description 1
- 229940104302 cytosine Drugs 0.000 description 1
- 230000001086 cytosolic effect Effects 0.000 description 1
- 125000001295 dansyl group Chemical group [H]C1=C([H])C(N(C([H])([H])[H])C([H])([H])[H])=C2C([H])=C([H])C([H])=C(C2=C1[H])S(*)(=O)=O 0.000 description 1
- 238000006731 degradation reaction Methods 0.000 description 1
- 230000006866 deterioration Effects 0.000 description 1
- 238000002050 diffraction method Methods 0.000 description 1
- PGUYAANYCROBRT-UHFFFAOYSA-N dihydroxy-selanyl-selanylidene-lambda5-phosphane Chemical compound OP(O)([SeH])=[Se] PGUYAANYCROBRT-UHFFFAOYSA-N 0.000 description 1
- NAGJZTKCGNOGPW-UHFFFAOYSA-K dioxido-sulfanylidene-sulfido-$l^{5}-phosphane Chemical compound [O-]P([O-])([S-])=S NAGJZTKCGNOGPW-UHFFFAOYSA-K 0.000 description 1
- 239000001177 diphosphate Substances 0.000 description 1
- 230000009977 dual effect Effects 0.000 description 1
- 230000005672 electromagnetic field Effects 0.000 description 1
- 210000002889 endothelial cell Anatomy 0.000 description 1
- 108010048367 enhanced green fluorescent protein Proteins 0.000 description 1
- 238000007824 enzymatic assay Methods 0.000 description 1
- 230000002255 enzymatic effect Effects 0.000 description 1
- 210000002919 epithelial cell Anatomy 0.000 description 1
- 210000003743 erythrocyte Anatomy 0.000 description 1
- ZMMJGEGLRURXTF-UHFFFAOYSA-N ethidium bromide Chemical compound [Br-].C12=CC(N)=CC=C2C2=CC=C(N)C=C2[N+](CC)=C1C1=CC=CC=C1 ZMMJGEGLRURXTF-UHFFFAOYSA-N 0.000 description 1
- 229960005542 ethidium bromide Drugs 0.000 description 1
- 125000001495 ethyl group Chemical group [H]C([H])([H])C([H])([H])* 0.000 description 1
- 238000000605 extraction Methods 0.000 description 1
- GVEPBJHOBDJJJI-UHFFFAOYSA-N fluoranthrene Natural products C1=CC(C2=CC=CC=C22)=C3C2=CC=CC3=C1 GVEPBJHOBDJJJI-UHFFFAOYSA-N 0.000 description 1
- GNBHRKFJIUUOQI-UHFFFAOYSA-N fluorescein Chemical compound O1C(=O)C2=CC=CC=C2C21C1=CC=C(O)C=C1OC1=CC(O)=CC=C21 GNBHRKFJIUUOQI-UHFFFAOYSA-N 0.000 description 1
- GTQFZXYECNSNNC-UHFFFAOYSA-N fluorescein 6-isothiocyanate Chemical compound O1C(=O)C2=CC=C(N=C=S)C=C2C21C1=CC=C(O)C=C1OC1=CC(O)=CC=C21 GTQFZXYECNSNNC-UHFFFAOYSA-N 0.000 description 1
- 238000000799 fluorescence microscopy Methods 0.000 description 1
- 238000001506 fluorescence spectroscopy Methods 0.000 description 1
- 108091006047 fluorescent proteins Proteins 0.000 description 1
- 102000034287 fluorescent proteins Human genes 0.000 description 1
- 238000007672 fourth generation sequencing Methods 0.000 description 1
- 238000013467 fragmentation Methods 0.000 description 1
- 238000006062 fragmentation reaction Methods 0.000 description 1
- 102000038383 gamma-secretases Human genes 0.000 description 1
- 108091007739 gamma-secretases Proteins 0.000 description 1
- 239000000499 gel Substances 0.000 description 1
- 230000030279 gene silencing Effects 0.000 description 1
- 238000012226 gene silencing method Methods 0.000 description 1
- 238000007903 genomic in situ hybridization Methods 0.000 description 1
- 210000004602 germ cell Anatomy 0.000 description 1
- PCHJSUWPFVWCPO-UHFFFAOYSA-N gold Chemical compound [Au] PCHJSUWPFVWCPO-UHFFFAOYSA-N 0.000 description 1
- 239000010931 gold Substances 0.000 description 1
- 229910052737 gold Inorganic materials 0.000 description 1
- 229940076085 gold Drugs 0.000 description 1
- 239000003102 growth factor Substances 0.000 description 1
- 238000004128 high performance liquid chromatography Methods 0.000 description 1
- 238000000589 high-performance liquid chromatography-mass spectrometry Methods 0.000 description 1
- 210000003630 histaminocyte Anatomy 0.000 description 1
- 239000005556 hormone Substances 0.000 description 1
- 229940088597 hormone Drugs 0.000 description 1
- 238000007849 hot-start PCR Methods 0.000 description 1
- 210000004408 hybridoma Anatomy 0.000 description 1
- BHEPBYXIRTUNPN-UHFFFAOYSA-N hydridophosphorus(.) (triplet) Chemical compound [PH] BHEPBYXIRTUNPN-UHFFFAOYSA-N 0.000 description 1
- 125000002887 hydroxy group Chemical group [H]O* 0.000 description 1
- 238000003018 immunoassay Methods 0.000 description 1
- 230000002163 immunogen Effects 0.000 description 1
- 229940072221 immunoglobulins Drugs 0.000 description 1
- 238000001114 immunoprecipitation Methods 0.000 description 1
- 238000012296 in situ hybridization assay Methods 0.000 description 1
- 238000001727 in vivo Methods 0.000 description 1
- 230000006698 induction Effects 0.000 description 1
- 102000006495 integrins Human genes 0.000 description 1
- 108010044426 integrins Proteins 0.000 description 1
- 230000003993 interaction Effects 0.000 description 1
- 230000003834 intracellular effect Effects 0.000 description 1
- 230000005865 ionizing radiation Effects 0.000 description 1
- 150000002500 ions Chemical class 0.000 description 1
- 238000001155 isoelectric focusing Methods 0.000 description 1
- 238000012004 kinetic exclusion assay Methods 0.000 description 1
- 229940039696 lactobacillus Drugs 0.000 description 1
- 229910052747 lanthanoid Inorganic materials 0.000 description 1
- 150000002602 lanthanoids Chemical class 0.000 description 1
- QDLAGTHXVHQKRE-UHFFFAOYSA-N lichenxanthone Natural products COC1=CC(O)=C2C(=O)C3=C(C)C=C(OC)C=C3OC2=C1 QDLAGTHXVHQKRE-UHFFFAOYSA-N 0.000 description 1
- 230000000670 limiting effect Effects 0.000 description 1
- 238000004895 liquid chromatography mass spectrometry Methods 0.000 description 1
- 238000003670 luciferase enzyme activity assay Methods 0.000 description 1
- 235000019689 luncheon sausage Nutrition 0.000 description 1
- 210000004698 lymphocyte Anatomy 0.000 description 1
- HAVFFEMDLROBGI-UHFFFAOYSA-N m8926c7ilx Chemical compound C1CC(O)CCN1CC1=CC=C(OC=2C3=C(C(NN=C33)=O)C=CC=2)C3=C1 HAVFFEMDLROBGI-UHFFFAOYSA-N 0.000 description 1
- 230000014759 maintenance of location Effects 0.000 description 1
- 238000004949 mass spectrometry Methods 0.000 description 1
- 230000007246 mechanism Effects 0.000 description 1
- 230000021121 meiosis Effects 0.000 description 1
- 102000006240 membrane receptors Human genes 0.000 description 1
- 108020004084 membrane receptors Proteins 0.000 description 1
- 229910052751 metal Inorganic materials 0.000 description 1
- 239000002184 metal Substances 0.000 description 1
- CXKWCBBOMKCUKX-UHFFFAOYSA-M methylene blue Chemical compound [Cl-].C1=CC(N(C)C)=CC2=[S+]C3=CC(N(C)C)=CC=C3N=C21 CXKWCBBOMKCUKX-UHFFFAOYSA-M 0.000 description 1
- 229960000907 methylthioninium chloride Drugs 0.000 description 1
- 238000000386 microscopy Methods 0.000 description 1
- 239000000203 mixture Substances 0.000 description 1
- TUGMVGKTLNQWJN-UHFFFAOYSA-N morpholin-4-ylmethylphosphonic acid Chemical class OP(O)(=O)CN1CCOCC1 TUGMVGKTLNQWJN-UHFFFAOYSA-N 0.000 description 1
- 239000013642 negative control Substances 0.000 description 1
- 238000007857 nested PCR Methods 0.000 description 1
- 230000004770 neurodegeneration Effects 0.000 description 1
- 208000015122 neurodegenerative disease Diseases 0.000 description 1
- 210000002682 neurofibrillary tangle Anatomy 0.000 description 1
- 230000016273 neuron death Effects 0.000 description 1
- 239000002858 neurotransmitter agent Substances 0.000 description 1
- PCHKPVIQAHNQLW-CQSZACIVSA-N niraparib Chemical compound N1=C2C(C(=O)N)=CC=CC2=CN1C(C=C1)=CC=C1[C@@H]1CCCNC1 PCHKPVIQAHNQLW-CQSZACIVSA-N 0.000 description 1
- 238000001821 nucleic acid purification Methods 0.000 description 1
- 125000003835 nucleoside group Chemical group 0.000 description 1
- FDLYAMZZIXQODN-UHFFFAOYSA-N olaparib Chemical compound FC1=CC=C(CC=2C3=CC=CC=C3C(=O)NN=2)C=C1C(=O)N(CC1)CCN1C(=O)C1CC1 FDLYAMZZIXQODN-UHFFFAOYSA-N 0.000 description 1
- 230000001590 oxidative effect Effects 0.000 description 1
- VYNDHICBIRRPFP-UHFFFAOYSA-N pacific blue Chemical compound FC1=C(O)C(F)=C2OC(=O)C(C(=O)O)=CC2=C1 VYNDHICBIRRPFP-UHFFFAOYSA-N 0.000 description 1
- 229940055729 papain Drugs 0.000 description 1
- 235000019834 papain Nutrition 0.000 description 1
- 230000036961 partial effect Effects 0.000 description 1
- 230000008506 pathogenesis Effects 0.000 description 1
- 229940111202 pepsin Drugs 0.000 description 1
- 238000012831 peritoneal equilibrium test Methods 0.000 description 1
- 125000002080 perylenyl group Chemical group C1(=CC=C2C=CC=C3C4=CC=CC5=CC=CC(C1=C23)=C45)* 0.000 description 1
- CSHWQDPOILHKBI-UHFFFAOYSA-N peryrene Natural products C1=CC(C2=CC=CC=3C2=C2C=CC=3)=C3C2=CC=CC3=C1 CSHWQDPOILHKBI-UHFFFAOYSA-N 0.000 description 1
- 239000010452 phosphate Substances 0.000 description 1
- 150000004713 phosphodiesters Chemical class 0.000 description 1
- 230000004962 physiological condition Effects 0.000 description 1
- 230000007505 plaque formation Effects 0.000 description 1
- 150000004032 porphyrins Chemical class 0.000 description 1
- 238000012636 positron electron tomography Methods 0.000 description 1
- 238000012877 positron emission topography Methods 0.000 description 1
- 229910052700 potassium Inorganic materials 0.000 description 1
- SCVFZCLFOSHCOH-UHFFFAOYSA-M potassium acetate Substances [K+].CC([O-])=O SCVFZCLFOSHCOH-UHFFFAOYSA-M 0.000 description 1
- 235000011056 potassium acetate Nutrition 0.000 description 1
- 238000001556 precipitation Methods 0.000 description 1
- 239000002243 precursor Substances 0.000 description 1
- 230000004952 protein activity Effects 0.000 description 1
- INCIMLINXXICKS-UHFFFAOYSA-M pyronin Y Chemical compound [Cl-].C1=CC(=[N+](C)C)C=C2OC3=CC(N(C)C)=CC=C3C=C21 INCIMLINXXICKS-UHFFFAOYSA-M 0.000 description 1
- 238000012175 pyrosequencing Methods 0.000 description 1
- WVIICGIFSIBFOG-UHFFFAOYSA-N pyrylium Chemical compound C1=CC=[O+]C=C1 WVIICGIFSIBFOG-UHFFFAOYSA-N 0.000 description 1
- 239000002096 quantum dot Substances 0.000 description 1
- 230000005855 radiation Effects 0.000 description 1
- 150000003254 radicals Chemical class 0.000 description 1
- 229910052761 rare earth metal Inorganic materials 0.000 description 1
- 150000002910 rare earth metals Chemical class 0.000 description 1
- 102000027426 receptor tyrosine kinases Human genes 0.000 description 1
- 108091008598 receptor tyrosine kinases Proteins 0.000 description 1
- 230000001603 reducing effect Effects 0.000 description 1
- 230000010076 replication Effects 0.000 description 1
- 239000011347 resin Substances 0.000 description 1
- 229920005989 resin Polymers 0.000 description 1
- 238000007894 restriction fragment length polymorphism technique Methods 0.000 description 1
- 230000001177 retroviral effect Effects 0.000 description 1
- MUSLHCJRTRQOSP-UHFFFAOYSA-N rhodamine 101 Chemical compound [O-]C(=O)C1=CC=CC=C1C(C1=CC=2CCCN3CCCC(C=23)=C1O1)=C2C1=C(CCC1)C3=[N+]1CCCC3=C2 MUSLHCJRTRQOSP-UHFFFAOYSA-N 0.000 description 1
- 239000001022 rhodamine dye Substances 0.000 description 1
- 239000002336 ribonucleotide Substances 0.000 description 1
- 108020004418 ribosomal RNA Proteins 0.000 description 1
- 125000000548 ribosyl group Chemical group C1([C@H](O)[C@H](O)[C@H](O1)CO)* 0.000 description 1
- YYMBJDOZVAITBP-UHFFFAOYSA-N rubrene Chemical compound C1=CC=CC=C1C(C1=C(C=2C=CC=CC=2)C2=CC=CC=C2C(C=2C=CC=CC=2)=C11)=C(C=CC=C2)C2=C1C1=CC=CC=C1 YYMBJDOZVAITBP-UHFFFAOYSA-N 0.000 description 1
- 150000003839 salts Chemical class 0.000 description 1
- 238000007480 sanger sequencing Methods 0.000 description 1
- JRPHGDYSKGJTKZ-UHFFFAOYSA-K selenophosphate Chemical compound [O-]P([O-])([O-])=[Se] JRPHGDYSKGJTKZ-UHFFFAOYSA-K 0.000 description 1
- 239000004065 semiconductor Substances 0.000 description 1
- 238000007841 sequencing by ligation Methods 0.000 description 1
- 238000007860 single-cell PCR Methods 0.000 description 1
- 229910052708 sodium Inorganic materials 0.000 description 1
- 239000001632 sodium acetate Substances 0.000 description 1
- 235000017281 sodium acetate Nutrition 0.000 description 1
- 230000000392 somatic effect Effects 0.000 description 1
- 125000006850 spacer group Chemical group 0.000 description 1
- 238000004611 spectroscopical analysis Methods 0.000 description 1
- PWEBUXCTKOWPCW-UHFFFAOYSA-L squarate Chemical compound [O-]C1=C([O-])C(=O)C1=O PWEBUXCTKOWPCW-UHFFFAOYSA-L 0.000 description 1
- 238000010186 staining Methods 0.000 description 1
- PJANXHGTPQOBST-UHFFFAOYSA-N stilbene Chemical compound C=1C=CC=CC=1C=CC1=CC=CC=C1 PJANXHGTPQOBST-UHFFFAOYSA-N 0.000 description 1
- 235000021286 stilbenes Nutrition 0.000 description 1
- 238000003860 storage Methods 0.000 description 1
- YBBRCQOCSYXUOC-UHFFFAOYSA-N sulfuryl dichloride Chemical compound ClS(Cl)(=O)=O YBBRCQOCSYXUOC-UHFFFAOYSA-N 0.000 description 1
- 230000007470 synaptic degeneration Effects 0.000 description 1
- 238000003786 synthesis reaction Methods 0.000 description 1
- 125000003698 tetramethyl group Chemical group [H]C([H])([H])* 0.000 description 1
- 125000003396 thiol group Chemical group [H]S* 0.000 description 1
- 229940113082 thymine Drugs 0.000 description 1
- 230000002103 transcriptional effect Effects 0.000 description 1
- 238000001890 transfection Methods 0.000 description 1
- 230000009261 transgenic effect Effects 0.000 description 1
- GWBUNZLLLLDXMD-UHFFFAOYSA-H tricopper;dicarbonate;dihydroxide Chemical compound [OH-].[OH-].[Cu+2].[Cu+2].[Cu+2].[O-]C([O-])=O.[O-]C([O-])=O GWBUNZLLLLDXMD-UHFFFAOYSA-H 0.000 description 1
- LENZDBCJOHFCAS-UHFFFAOYSA-N tris Chemical compound OCC(N)(CO)CO LENZDBCJOHFCAS-UHFFFAOYSA-N 0.000 description 1
- 238000002604 ultrasonography Methods 0.000 description 1
- ORHBXUUXSCNDEV-UHFFFAOYSA-N umbelliferone Chemical compound C1=CC(=O)OC2=CC(O)=CC=C21 ORHBXUUXSCNDEV-UHFFFAOYSA-N 0.000 description 1
- HFTAFOQKODTIJY-UHFFFAOYSA-N umbelliferone Natural products Cc1cc2C=CC(=O)Oc2cc1OCC=CC(C)(C)O HFTAFOQKODTIJY-UHFFFAOYSA-N 0.000 description 1
- 229940035893 uracil Drugs 0.000 description 1
- 229950011257 veliparib Drugs 0.000 description 1
- JNAHVYVRKWKWKQ-CYBMUJFWSA-N veliparib Chemical compound N=1C2=CC=CC(C(N)=O)=C2NC=1[C@@]1(C)CCCN1 JNAHVYVRKWKWKQ-CYBMUJFWSA-N 0.000 description 1
- 238000011179 visual inspection Methods 0.000 description 1
- 239000011782 vitamin Substances 0.000 description 1
- 235000013343 vitamin Nutrition 0.000 description 1
- 229940088594 vitamin Drugs 0.000 description 1
- 229930003231 vitamin Natural products 0.000 description 1
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 1
- QDLHCMPXEPAAMD-QAIWCSMKSA-N wortmannin Chemical compound C1([C@]2(C)C3=C(C4=O)OC=C3C(=O)O[C@@H]2COC)=C4[C@@H]2CCC(=O)[C@@]2(C)C[C@H]1OC(C)=O QDLHCMPXEPAAMD-QAIWCSMKSA-N 0.000 description 1
- QDLHCMPXEPAAMD-UHFFFAOYSA-N wortmannin Natural products COCC1OC(=O)C2=COC(C3=O)=C2C1(C)C1=C3C2CCC(=O)C2(C)CC1OC(C)=O QDLHCMPXEPAAMD-UHFFFAOYSA-N 0.000 description 1
- 150000003732 xanthenes Chemical class 0.000 description 1
- 229940075420 xanthine Drugs 0.000 description 1
Images
Classifications
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12Q—MEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
- C12Q1/00—Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
- C12Q1/68—Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
- C12Q1/6876—Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
- C12Q1/6883—Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/46—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
- C07K14/47—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
- C07K14/4701—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
- C07K14/4702—Regulators; Modulating activity
- C07K14/4703—Inhibitors; Suppressors
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/46—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
- C07K14/47—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
- C07K14/4701—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
- C07K14/4711—Alzheimer's disease; Amyloid plaque core protein
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/11—DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
- C12N15/113—Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2310/00—Structure or type of the nucleic acid
- C12N2310/10—Type of nucleic acid
- C12N2310/12—Type of nucleic acid catalytic nucleic acids, e.g. ribozymes
- C12N2310/122—Hairpin
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2310/00—Structure or type of the nucleic acid
- C12N2310/10—Type of nucleic acid
- C12N2310/14—Type of nucleic acid interfering N.A.
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12Q—MEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
- C12Q2600/00—Oligonucleotides characterized by their use
- C12Q2600/156—Polymorphic or mutational markers
Definitions
- the one or more non-classical variant(s) comprises a portion or all of exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof.
- the one or more non-classical variant(s) does not comprise exon 8.
- one or more exon(s) in the one or more non-classical variant(s) is rearranged as compared to a control.
- the one or more non-classical variant(s) comprises a single nucleotide variation (SNV) in APP.
- the SNV in APP translates to amino acid positions in APP selected from a group consisting of A673V, A713T, T714I, V715M, V715A, I716M, V717I, V717F, T719P, and L723P.
- generation of the one or more non-classical variant(s) involves transcription.
- the one or more non-classical variant(s) comprises a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16.
- the disease or disorder is Alzheimer's disease.
- Alzheimer's disease is familial Alzheimer's disease (FAD) or sporadic Alzheimer's disease.
- the agent that inhibits the activity of APP is an antibody, an antigen binding fragment, a RNA interfering agent (RNAi), a small interfering RNA (siRNA), a short hairpin RNA (shRNA), a microRNA (miRNA), an antisense oligonucleotide, a peptide, a peptidomimetic, a small molecule, or an aptamer.
- the antibody binds to a protein encoded by the one or more non-classical variant(s).
- the antisense oligonucleotide targets RNA or DNA of APP.
- the RNA is mRNA.
- the DNA is genomic DNA.
- the protein is encoded by the one or more non-classical variant(s) comprising a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16.
- the agent does not inhibit activity of a wild-type amyloid beta precursor protein gene or protein thereof.
- the unwanted accumulation of amyloid beta protein is plaque depositions comprising a portion of or all amyloid beta protein, or accumulation of soluble amyloid beta protein.
- the unwanted accumulation of amyloid beta protein results from increased APP associated with the one or more non-classical variant(s) of APP gene.
- kits for diagnosing and treating a disease or disorder characterized by unwanted accumulation of amyloid beta protein in an individual in need thereof comprising:
- kits for diagnosing and treating a disease or disorder characterized by unwanted accumulation of amyloid beta protein in an individual in need thereof comprising:
- detecting one or more non-classical variant(s) of an APP gene in an individual in need thereof comprising: detecting an expression profile of the one or more non-classical variant(s) of the APP gene in a biological sample from the individual by a method comprising long-read sequencing of the biological sample.
- the expression profile is expression level of the one or more non-classical variant(s).
- the expression profile is expression of a set of different non-classical variants.
- the one or more non-classical variant(s) comprises a portion or all of exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof.
- the one or more non-classical variant(s) does not comprise exon 8. In some embodiments, one or more exon(s) in the one or more non-classical variant(s) is rearranged as compared to a control. In some embodiments, the one or more non-classical variant(s) comprises a single nucleotide variation (SNV) in APP. In some embodiments, the SNV in APP translates to amino acid positions in APP selected from a group consisting of A673V, A713T, T714I, V715M, V715A, I716M, V717I, V717F, T719P, and L723P.
- SNV single nucleotide variation
- generation of the one or more non-classical variant(s) involves transcription.
- the one or more non-classical variant(s) comprises a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16.
- the expression profile of the one or more non-classical variant(s) is associated with a disease or disorder characterized by unwanted accumulation of amyloid beta protein.
- the unwanted accumulation of amyloid beta protein is plaque depositions comprising a portion of or all amyloid beta protein, or accumulation of soluble amyloid beta protein.
- the expression profile of the one or more non-classical variant(s) is associated with unwanted accumulation of amyloid beta protein, and wherein the individual does not have a disease or disorder.
- the disease or disorder is Alzheimer's disease.
- the biological sample is collected from blood or cerebrospinal fluid.
- the biological sample comprises RNA or DNA.
- the DNA is genomic DNA, extrachromosomal DNA, or circular DNA.
- the DNA is genomic DNA.
- methods further comprise isolating and purifying RNA or DNA from the biological sample prior to the long-read sequencing.
- methods further comprise reverse transcribing RNA to cDNA prior to the long-read sequencing.
- an average read length for the long-read sequencing is at least 5000 bases.
- the long-read sequencing is RNA sequencing (RNA-seq).
- the long-read sequencing is DNA sequencing.
- detecting the expression profile comprises quantifying the expression level of the one or more non-classical variant(s) by a method comprising counting a number of reads that map to a sequence of the one or more non-classical variant(s).
- the individual is suspected of having or being predisposed to Alzheimer's disease.
- detecting one or more non-classical variant(s) of an APP gene in an individual in need thereof comprising: detecting an expression profile or an activity profile of the one or more non-classical variant of the APP gene in a biological sample from the individual by a method comprising binding of one or more probe(s) to the one or more non-classical variant(s).
- the expression profile is expression level of the one or more non-classical variant(s).
- the activity profile is activity level of the one or more non-classical variant(s).
- the expression profile is expression of a set of different non-classical variants.
- the activity profile is activity of a set of different non-classical variants.
- detecting the expression profile comprises quantifying expression level by a method comprising quantitative polymerase chain reaction (qPCR).
- the one or more non-classical variant(s) comprises a portion or all of exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof.
- the one or more non-classical variant(s) does not comprise exon 8.
- one or more exon(s) in the one or more non-classical variant(s) is rearranged as compared to a control.
- the one or more non-classical variant(s) comprises a single nucleotide variation (SNV) in APP.
- the SNV in APP translates to amino acid positions in APP selected from a group consisting of A673V, A713T, T714I, V715M, V715A, I716M, V717I, V717F, T719P, and L723P.
- generation of the one or more non-classical variant(s) involves transcription.
- the one or more non-classical variant(s) comprises a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16.
- the expression profile or the activity level of the one or more non-classical variant(s) is associated with a disease or disorder characterized by unwanted accumulation of amyloid beta protein.
- the unwanted accumulation of amyloid beta protein is plaque depositions comprising a portion of or all amyloid beta protein, or accumulation of soluble amyloid beta protein.
- the expression profile or the activity profile of the one or more non-classical variant(s) is associated with unwanted accumulation of amyloid beta protein, and wherein the individual does not have a disease or disorder.
- the disease or disorder is Alzheimer's disease.
- the biological sample is collected from blood or cerebrospinal fluid.
- the biological sample comprises RNA, DNA, or protein.
- the DNA is genomic DNA, extrachromosomal DNA, or circular DNA. In some embodiments, the DNA is genomic DNA.
- the one or more probe(s) are selected from polynucleotides or polypeptides. In some embodiments, the one or more probe(s) hybridize to RNA or DNA within the biological sample. In some embodiments, the one or more probe(s) hybridize to a range of about 35 to about 50 nucleotides of the RNA or DNA. In some embodiments, methods further comprise capturing the one or more non-classical variant(s) from the biological sample on a solid support prior to contacting the one or more non-classical variant with the one or more probe(s).
- the one or more non-classical variant(s) is detected by in situ hybridization or immunological hybridization.
- the in situ hybridization is chromogenic in situ hybridization or fluorescence in situ hybridization.
- detecting binding of the one or more probe(s) to the one or more non-classical variant(s) further comprises a pull-down assay.
- the one or more probe(s) for the pull-down assay are designed to hybridize to a portion or all of exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof.
- a probe in the one or more probe(s) is labeled using an affinity tag.
- the affinity tag is biotin, desthiobiotin, histidine, polyhistidine, myc, hemagglutinin (HA), FLAG, glutathione S transferase (GST), or derivatives thereof.
- the affinity tag is recognized by avidin, streptavidin, nickel, or glutathione.
- the one or more probe(s) bind to one or more protein(s) encoded by the one or more non-classical variant(s).
- the one or more probe(s) comprise an antibody or fragment thereof.
- a probe in the one or more probe(s) is labeled using a fluorochrome or a radioactive isotope.
- a disease or disorder in an individual characterized by unwanted accumulation of amyloid beta protein comprising: identifying the individual as having the disease or disorder characterized by unwanted accumulation of amyloid beta protein by comparing an expression profile of one or more non-classical variant(s) of an APP gene to a reference expression profile of the one or more non-classical variant(s) derived from a cohort of control individuals, wherein the expression profile of the one or more non-classical variant(s) is measured by a method comprising long-read sequencing of a biological sample from the individual; and wherein the expression profile of the one or more non-classical variant(s) is associated with the disease or disorder.
- the expression profile is expression level of the one or more non-classical variant(s). In some embodiments, the expression profile is expression of a set of different non-classical variants.
- the long-read sequencing is RNA sequencing (RNA-seq). In some embodiments, the long-read sequencing is DNA sequencing.
- methods further comprise quantifying the expression level of the one or more non-classical variant(s) by a method comprising counting a number of reads that map to a sequence of the one or more non-classical variant(s). In some embodiments, the one or more non-classical variant(s) comprises a portion or all of exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof.
- the one or more non-classical variant(s) does not comprise exon 8. In some embodiments, one or more exon(s) in the one or more non-classical variant(s) is rearranged as compared to a control. In some embodiments, the one or more non-classical variant(s) comprises a single nucleotide variation (SNV) in APP. In some embodiments, the SNV in APP translates to amino acid positions in APP selected from a group consisting of A673V, A713T, T714I, V715M, V715A, I716M, V717I, V717F, T719P, and L723P.
- SNV single nucleotide variation
- generation of the one or more non-classical variant(s) involves transcription.
- the one or more non-classical variant(s) comprises a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16.
- the disease or disorder is Alzheimer's disease.
- Alzheimer's disease is familial Alzheimer's disease (FAD) or sporadic Alzheimer's disease.
- the individual is suspected of having or being predisposed to Alzheimer's disease.
- the biological sample is collected from blood or cerebrospinal fluid.
- methods further comprise isolating and purifying RNA or DNA from the biological sample prior to the long-read sequencing.
- methods further comprise reverse transcribing RNA to cDNA prior to the long-read sequencing.
- an average read length for the long-read sequencing is at least 5000 bases.
- the unwanted accumulation of amyloid beta protein is plaque depositions comprising a portion of or all amyloid beta protein, or accumulation of soluble amyloid beta protein.
- a disease or disorder in an individual characterized by unwanted accumulation of amyloid beta protein comprising: identifying the individual as having the disease or disorder characterized by unwanted accumulation of amyloid beta protein by comparing an expression profile or an activity profile of one or more non-classical variant(s) of an APP gene to a reference expression profile of the one or more non-classical variant(s) derived from a cohort of control individuals, wherein the expression profile or the activity profile of the one or more non-classical variant(s) is measured by a method comprising binding of one or more probe(s) to a biological sample from the individual; and wherein the expression profile or the activity profile of the one or more non-classical variant(s) is associated with the disease or disorder.
- the expression profile is expression level of the one or more non-classical variant(s). In some embodiments, the activity profile is activity level of the one or more non-classical variant(s). In some embodiments, the expression profile is expression of a set of different non-classical variants. In some embodiments, the activity profile is activity of a set of different non-classical variants. In some embodiments, methods further comprise quantifying the expression level by a method comprising quantitative polymerase chain reaction (qPCR). In some embodiments, methods further comprise capturing the one or more non-classical variant(s) from the biological sample on a solid support prior to contacting the one or more non-classical variant(s) with the one or more probe(s).
- qPCR quantitative polymerase chain reaction
- the one or more non-classical variant(s) is detected by in situ hybridization or immunological hybridization.
- the in situ hybridization is chromogenic in situ hybridization or fluorescence in situ hybridization.
- binding of the one or more probe(s) to the one or more non-classical variant(s) further comprises a pull-down assay.
- the one or more probe(s) for the pull-down assay are designed to hybridize to a portion or all of exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof.
- a probe in the one or more probe(s) is labeled using an affinity tag.
- the affinity tag is biotin, desthiobiotin, histidine, polyhistidine, myc, hemagglutinin (HA), FLAG, glutathione S transferase (GST), or derivatives thereof.
- the affinity tag is recognized by avidin, streptavidin, nickel, or glutathione.
- the one or more probe(s) are selected from polynucleotides or polypeptides.
- the one or more probe(s) hybridize to RNA or DNA within the biological sample. In some embodiments, the one or more probe(s) hybridize to a range of about 35 to about 50 nucleotides of the RNA or DNA.
- the one or more probe(s) bind to one or more protein(s) encoded by the one or more non-classical variant(s).
- the one or more probe(s) comprise an antibody or fragment thereof.
- a probe in the one or more probe(s) is labeled using a fluorochrome or a radioactive isotope.
- the unwanted accumulation of amyloid beta protein is plaque depositions comprising amyloid beta protein, or accumulation of soluble amyloid beta protein.
- kits for detecting one or more non-classical variant(s) of an APP gene in a biological sample of an individual comprising:
- in vitro methods of screening for a therapeutic agent for treating a disease or disorder characterized by unwanted accumulation of amyloid beta protein comprising:
- in vitro methods of screening for a therapeutic agent for treating a disease or disorder characterized by unwanted accumulation of amyloid beta protein comprising:
- in vitro methods of screening for a therapeutic agent for treating a disease or disorder characterized by unwanted accumulation of amyloid beta protein comprising:
- identifying one or more non-classical variant(s) of an APP gene comprising:
- therapeutic agents identified by any of the methods described herein.
- a disease or disorder characterized by unwanted accumulation of amyloid beta protein in an individual in need thereof comprising: administering to the individual an agent that inhibits generation of one or more non-classical variant(s) of an APP gene.
- the generation of the one or more non-classical variant(s) involves transcription.
- the agent edits RNA.
- the agent edits DNA.
- the one or more non-classical variant(s) comprises a portion or all of exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof.
- the one or more non-classical variant(s) comprises a single nucleotide variation (SNV) in APP.
- SNV single nucleotide variation
- the SNV in APP translates to amino acid positions in APP selected from a group consisting of A673V, A713T, T714I, V715M, V715A, I716M, V717I, V717F, T719P, and L723P.
- the one or more non-classical variant(s) does not comprise exon 8.
- one or more exon(s) in the one or more non-classical variant(s) is rearranged as compared to a control.
- the one or more non-classical variant(s) comprises a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16.
- the disease or disorder is Alzheimer's disease.
- Alzheimer's disease is familial Alzheimer's disease (FAD) or sporadic Alzheimer's disease.
- the unwanted accumulation of amyloid beta protein is plaque depositions comprising a portion of or all amyloid beta protein, or accumulation of soluble amyloid beta protein.
- the unwanted accumulation of amyloid beta protein results from increased APP associated with the one or more non-classical variant(s) of APP gene.
- a composition comprising: (a) a nuclease; and (b) a guide nucleic acid comprising a region that is complementary to a target nucleic acid, wherein the target nucleic acid is a non-classical variant of APP.
- the nuclease is a CRISPR-associated protein (Cas).
- the Cas protein is Cas9. In some embodiments, the Cas protein is Cas13.
- the target nucleic acid is RNA of the non-classical variant of APP. In some embodiments, the target nucleic acid is DNA of the non-classical variant of APP. In some embodiments, the non-classical variant comprises a portion or all of exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof. In some embodiments, the non-classical variant comprises a single nucleotide variation (SNV) in APP.
- SNV single nucleotide variation
- the SNV in APP translates to amino acid positions in APP selected from a group consisting of A673V, A713T, T714I, V715M, V715A, I716M, V717I, V717F, T719P, and L723P.
- the non-classical variant does not comprise exon 8.
- one or more exon(s) in the non-classical variant is rearranged as compared to a control.
- the non-classical variant comprises a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16.
- the disease or disorder is Alzheimer's disease.
- Alzheimer's disease is familial Alzheimer's disease (FAD) or sporadic Alzheimer's disease.
- the unwanted accumulation of amyloid beta protein is plaque depositions comprising a portion of or all amyloid beta protein, or accumulation of soluble amyloid beta protein.
- unwanted accumulation of amyloid beta protein results from increased APP associated with the non-classical variant of APP gene.
- methods method of detecting one or more non-classical variant(s) of an APP gene in an individual in need thereof comprising: detecting an expression profile of the one or more non-classical variant(s) of the APP gene in a biological sample from the individual by (a) long-read sequencing of RNA or DNA encoding for the one or more non-classical variant(s), or (b) binding of one or more probe(s) to the one or more non-classical variant(s).
- the expression profile is expression level of the one or more non-classical variant(s) or expression of a set of different non-classical variants.
- the one or more non-classical variant(s) comprises a portion or all of exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof. In some embodiments, the one or more non-classical variant(s) does not comprise exon 8. In some embodiments, one or more exon(s) in the one or more non-classical variant(s) is rearranged as compared to a control. In some embodiments, the one or more non-classical variant(s) comprises a single nucleotide variation (SNV) in APP.
- SNV single nucleotide variation
- the SNV in APP translates to amino acid positions in APP selected from a group consisting of A673V, A713T, T714I, V715M, V715A, I716M, V717I, V717F, T719P, and L723P.
- the one or more non-classical variant(s) comprises a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16.
- the expression profile of the one or more non-classical variant(s) is associated with a disease or disorder characterized by unwanted accumulation of amyloid beta protein.
- the unwanted accumulation of amyloid beta protein is plaque depositions comprising a portion of or all amyloid beta protein, or accumulation of soluble amyloid beta protein.
- the expression profile of the one or more non-classical variant(s) is associated with unwanted accumulation of amyloid beta protein, and wherein the individual does not have a disease or disorder.
- the biological sample is blood or cerebrospinal fluid.
- the DNA is genomic DNA, extrachromosomal DNA, or circular DNA.
- detecting the expression profile comprises quantifying the expression level of the one or more non-classical variant(s) by a method comprising counting a number of reads that map to a sequence of the one or more non-classical variant(s).
- the individual is suspected of having or being predisposed to Alzheimer's disease.
- the one or more probe(s) hybridize to RNA or DNA encoding for the one or more non-classical variant(s) within the biological sample.
- the one or more probe(s) hybridize to an intraexonic junction between exon 1 and exon 11, exon 1 and exon 14, exon 2 and exon 17, exon 2 and exon 14, exon 2 and exon 18, exon 2 and exon 16, exon 3 and exon 16, exon 3 and exon 14, exon 3 and exon 17, exon 6 and exon 17, exon 6 and exon 18, exon 3 and exon 9, exon 2 and exon 9, exon 16 and exon 18, exon 6 and exon 12, exon 5 and exon 16, or exon 16 and exon 17.
- the one or more non-classical variant(s) is detected by in situ hybridization or immunological hybridization. In some embodiments, detecting binding of the one or more probe(s) to the one or more non-classical variant(s) further comprises a pull-down assay. In some embodiments, the one or more probe(s) for the pull-down assay are designed to hybridize to a portion or all of exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof. In some embodiments, a probe in the one or more probe(s) is labeled using an affinity tag. In some embodiments, the one or more probe(s) bind to one or more protein(s) encoded by the one or more non-classical variant(s).
- the one or more probe(s) comprise an antibody or fragment thereof.
- a probe in the one or more probe(s) is labeled using a fluorochrome or a radioactive isotope.
- methods further comprise administering an agent that inhibits activity of the one or more non-classical variant(s).
- the agent that inhibits the activity of APP is an antibody, an antigen binding fragment, a RNA interfering agent (RNAi), a small interfering RNA (siRNA), a short hairpin RNA (shRNA), a microRNA (miRNA), an antisense oligonucleotide, a peptide, a peptidomimetic, a small molecule, or an aptamer.
- RNAi RNA interfering agent
- siRNA small interfering RNA
- shRNA short hairpin RNA
- miRNA microRNA
- an antisense oligonucleotide targets RNA or DNA of APP.
- methods further comprise administering a cholinesterase inhibitor, N-methyl-D-aspartate (NMDA) receptor antagonist, or an anti-amyloid beta antibody.
- NMDA N-methyl-D-aspartate
- the cholinesterase inhibitor is selected from the group consisting of Donepezil, Galantamine, and Rivastigmine.
- the NMDA receptor antagonist is memantine.
- the anti-amyloid beta antibody is selected from the group consisting of Bapineuzumab, Solanezumab, Gantenerumab, Crenezumab, BAN2401, Ponezumab, and Aducanumab.
- the expression profile is expression level of the one or more non-classical variant(s), and wherein the activity profile is activity level of the one or more non-classical variant(s). In some embodiments, the expression profile is expression of a set of different non-classical variants, and wherein the activity profile is activity of a set of different non-classical variants. In some embodiments, methods further comprise measuring the expression profile by a method comprising long-read sequencing of RNA or DNA from the biological sample from the individual, wherein the RNA or DNA encodes the one or more non-classical variant(s).
- measuring the expression profile comprises quantifying the expression level of the one or more non-classical variant(s) by a method comprising counting a number of reads that map to a sequence of the one or more non-classical variant(s).
- the one or more non-classical variant(s) comprises a portion or all of exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof.
- the one or more non-classical variant(s) does not comprise exon 8.
- one or more exon(s) in the one or more non-classical variant(s) is rearranged as compared to a control.
- the one or more non-classical variant(s) comprises a single nucleotide variation (SNV) in APP.
- the SNV in APP translates to amino acid positions in APP selected from a group consisting of A673V, A713T, T714I, V715M, V715A, I716M, V717I, V717F, T719P, and L723P.
- the one or more non-classical variant(s) comprises a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16.
- the disease or disorder is Alzheimer's disease.
- the Alzheimer's disease is familial Alzheimer's disease (FAD) or sporadic Alzheimer's disease.
- the biological sample is blood or cerebrospinal fluid.
- the biological sample comprises RNA, DNA, or protein encoding for the one or more non-classical variant(s).
- the DNA is genomic DNA, extrachromosomal DNA, or circular DNA.
- measuring the expression profile comprises a method comprising binding of one or more probe(s) to the one or more non-classical variant(s).
- the one or more probe(s) hybridize to an intraexonic junction between exon 1 and exon 11, exon 1 and exon 14, exon 2 and exon 17, exon 2 and exon 14, exon 2 and exon 18, exon 2 and exon 16, exon 3 and exon 16, exon 3 and exon 14, exon 3 and exon 17, exon 6 and exon 17, exon 6 and exon 18, exon 3 and exon 9, exon 2 and exon 9, exon 16 and exon 18, exon 6 and exon 12, exon 5 and exon 16, or exon 16 and exon 17.
- measuring the expression profile comprises quantifying the expression level by a method comprising quantitative polymerase chain reaction (qPCR).
- the one or more probe(s) hybridize to RNA or DNA encoding for the one or more non-classical variant(s) within the biological sample.
- the one or more non-classical variant(s) is detected by in situ hybridization or immunological hybridization.
- measuring binding of the one or more probe(s) to the one or more non-classical variant(s) further comprises a pull-down assay.
- the one or more probe(s) for the pull-down assay are designed to hybridize to a portion or all of exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof.
- a probe of the one or more probe(s) is labeled using an affinity tag.
- the one or more probe(s) bind to one or more protein(s) encoded by the one or more non-classical variant(s).
- the one or more probe(s) comprise an antibody or fragment thereof.
- a probe of the one or more probe(s) is labeled using a fluorochrome or a radioactive isotope.
- the unwanted accumulation of amyloid beta protein is plaque depositions comprising a portion of or all amyloid beta protein, or accumulation of soluble amyloid beta protein.
- methods further comprise administering an agent that inhibits activity of the one or more non-classical variant(s).
- the agent that inhibits the activity of APP is an antibody, an antigen binding fragment, a RNA interfering agent (RNAi), a small interfering RNA (siRNA), a short hairpin RNA (shRNA), a microRNA (miRNA), an antisense oligonucleotide, a peptide, a peptidomimetic, a small molecule, or an aptamer.
- RNAi RNA interfering agent
- siRNA small interfering RNA
- shRNA short hairpin RNA
- miRNA microRNA
- an antisense oligonucleotide targets RNA or DNA of APP.
- methods further comprise administering a cholinesterase inhibitor, N-methyl-D-aspartate (NMDA) receptor antagonist, or an anti-amyloid beta antibody.
- NMDA N-methyl-D-aspartate
- the cholinesterase inhibitor is selected from the group consisting of Donepezil, Galantamine, and Rivastigmine.
- the NMDA receptor antagonist is memantine.
- the anti-amyloid beta antibody is selected from the group consisting of Bapineuzumab, Solanezumab, Gantenerumab, Crenezumab, BAN2401, Ponezumab, and Aducanumab.
- a disease or disorder characterized by unwanted accumulation of amyloid beta protein in an individual in need thereof comprising:
- kits for detecting one or more non-classical variant(s) of an APP gene in a biological sample of an individual comprising:
- FIG. 1 illustrates a schema of amyloid precursor protein (APP) mRNA and cDNA.
- APP amyloid precursor protein
- FIG. 2 illustrates a gel electrophoresis of RT-PCR from neurons isolated from cortices (CTX) and cerebellums (CBL) from non-diseased (Non-AD) postmortem brains and Alzheimer's disease (AD) brains.
- CX cortices
- CBL cerebellums
- Non-AD non-diseased postmortem brains
- AD Alzheimer's disease
- FIG. 3 illustrates a Southern blot of RT-PCR from neurons isolated from cortices (CTX) and cerebellums (CBL) from non-diseased (Non-AD) postmortem brains and Alzheimer's disease (AD) brains hybridized with APP cDNA probe.
- CX cortices
- CBL cerebellums
- P and N represent positive (APP plasmid) and negative (Presenilin 1 plasmid) control, respectively.
- FIG. 4 illustrates a gel electrophoresis of RT-PCR from neurons isolated from cortices (CTX) and cerebellums (CBL) from non-diseased (Non-AD) postmortem brains and Alzheimer's disease (AD) brains. Arrows indicate positive signals corresponding to signal from Southern blot.
- FIG. 5 illustrates exemplary non-classical variants.
- FIG. 6A illustrates amyloid beta positive clones from non-diseased (Non-AD) brains and Alzheimer's disease (AD) brains detected with amyloid beta (A ⁇ ) probes and APP cDNA probes.
- FIG. 6B illustrates a graph of percentage of amyloid beta in non-diseased (Non-AD) brains and Alzheimer's disease (AD) brains.
- FIG. 7 illustrates a gel electrophoresis of RT-PCR from frontal cortices of Alzheimer's disease (AD) brains (AD1 and AD3) in sorted neuronal and non-neuronal nuclei and bulk RNA.
- AD Alzheimer's disease
- FIG. 8A illustrates a gel electrophoresis of PCR for genomic DNA isolated from frontal cortices of non-diseased brains and Alzheimer's disease (AD) brains (AD1 and AD3) with APP primers.
- NTC represents no template control.
- FIG. 8B illustrates a gel electrophoresis of PCR for genomic DNA isolated from frontal cortices of non-diseased brains and Alzheimer's disease (AD) brains (AD1 and AD3) with Presenilin 1 (PSEN1) primers.
- NTC represents no template control
- PC represents positive control.
- FIG. 9 illustrates a graph of relative ⁇ CT for neuronal and non-neuronal samples in which cAPP-R3/16 and PSEN are detected.
- FIG. 10 illustrates a schema for detection of non-classical variants by DNA pull-down assay.
- FIG. 11 illustrates genomic rearrangements in an Alzheimer's disease (AD) neuronal nuclei detected by DNA in situ hybridization.
- AD Alzheimer's disease
- FIG. 12 illustrates intraexonic rearrangements in tissue sections from an Alzheimer's disease (AD) brain detected by RNA in situ hybridization of non-classical variant cAPP-R3/16 (red, 1201). Amyloid beta (A ⁇ ) is stained in brown (1203). Tissue sections are visualized at 200 ⁇ and 630 ⁇ magnification.
- AD Alzheimer's disease
- a ⁇ Amyloid beta
- FIG. 13 illustrates non-classical variants detected by single molecule real-time sequencing.
- FIG. 14 illustrates the presence of exon 16 and exon 17 junction in neuronal and non-neuronal nuclei from non-diseased and Alzheimer's disease (AD) brains detected by DNA in situ hybridization.
- AD Alzheimer's disease
- FIG. 15A illustrates immunofluorescence images of cells transfected with non-classical variants cAPP-R3/16, cAPP-R2/18, and ncAPP-R1/4 that are hemagglutinin (HA) epitope tagged (left panel). EGFP co-expression (middle panel) and DAPI staining (right panel) are shown.
- HA hemagglutinin
- FIG. 15B illustrates a Western blot of cell lysate from cells transfected with non-classical variants cAPP-R3/16, cAPP-R2/18, and ncAPP-R1/4 that are hemagglutinin (HA) epitope tagged.
- the Western blot was probed with HA antibody.
- FIG. 16A illustrates a gel electrophoresis of RT-PCR of LN-229 cells transfected with non-classical variant cAPP-R3/16 or wild-type APP (APP).
- FIG. 16B illustrates cAPP-R3/16 induction in LN-229 cells transfected with cAPP-R3/16 or wild-type APP (APP) detected by DNA in situ hybridization using cAPP-R3/16 probe and PPIB probe as a positive control.
- APP wild-type APP
- FIG. 17A illustrates a schematic for identification of non-classical RNA variants of APP from populations of neurons.
- 50-neuronal nuclei were sorted from human prefrontal cortices (FCTX) by fluorescence-activated nuclear sorting (FANS) and used for (2) RT-PCR. Resulting RT-PCR products were screened by (3) Southern blot with 32 P-labeled APP cDNA probes. (4) Bands with positive signals from duplicate gels were cloned and sequenced. (5) Non-classical variants were identified.
- Figure discloses SEQ ID NO: 33.
- FIG. 17B illustrates electrophoresis of RT-PCR products from 3 non-diseased and 3 sporadic AD brains.
- APP and PSEN1 plasmids were run as positive and negative controls for Southern blotting, respectively.
- FIG. 17C illustrates a gel electrophoresis of RT-PCR for bulk RNA detecting canonical APP splice variants as major products.
- Bulk RNA from 3 non-diseased and 3 sporadic Alzheimer's disease (SAD) prefrontal cortices was used for APP RT-PCR.
- Major products detected were canonical APP splice variants.
- FIG. 17D illustrates Southern blot of RT-PCR products. Arrows indicate examples of corresponding bands from FIG. 17B that were cloned and Sanger sequenced.
- FIG. 17E illustrates a schema of structure of human APP genomic locus.
- FIG. 17F illustrates schemas of non-classical RNA variants of APP identified by RT-PCR.
- FIG. 17G illustrates schemas of sequence homology of non-classical RNA variants of APP at intraexonic junctions. Homology sequences of proximal and distal exons are shaded in gray. Middle sequence is the identified variant, top and bottom sequences are publicly available coding sequences from NM 000484.3 from the respective exons. Nucleotide variations are indicated an asterisk. Non-classical RNA variants identified by Sanger sequencing and PacBio data sets are shown with R and P, respectively. Figure discloses SEQ ID NOS 34-42, respectively, in order of appearance.
- FIG. 17H illustrates non-classical RNA variants of APP identified from independent long-read single molecule real-time (SMRT) sequencing data sets.
- FIG. 18A illustrates a schema for analyzing non-classical genomic cDNA (gencDNA) variants of APP.
- GencDNA non-classical genomic cDNA
- FCTX human prefrontal cortices
- jgISH junction-specific genomic in situ hybridization
- FIG. 18B illustrates a gel electrophoresis of genomic DNA PCR products with APP and PSEN1 primer sets using DNA from non-diseased (ND) and sporadic AD (SAD) neurons.
- ND non-diseased
- SAD sporadic AD
- NC Non-template control
- PC positive control
- FIG. 18C illustrates a schema showing 13 variants identified first by RT-PCR (APP-R) and DNA PCR (APP-D). Seven were identified in both methods, five by RT-PCR only, and one by DNA PCR only.
- APP-R RT-PCR
- APP-D DNA PCR
- FIGS. 19A-19B illustrate jgISH performed with sense and anti-sense probes targeting APP exon 16 and exon 17 junction (Ex 16/17) ( FIG. 19A ) and intraexonic junction between APP exon 3 and exon 16 (IEJ 3/16) on SAD neuronal nuclei ( FIG. 19B ).
- FIGS. 19C-19D illustrate RNA-in situ hybridization with sense and antisense jgISH probes on human tissue sections.
- FIG. 19C shows Ex 16/17 sense and anti-sense probes for RNA-in situ hybridization on human prefrontal cortex tissue sections.
- FIG. 19D shows IEJ 3/16 sense and anti-sense probes for RNA-in situ hybridization on human prefrontal cortex tissue sections. Scale bars are 10 ⁇ m.
- FIGS. 19E-19J illustrate restriction enzyme (RE) digestion.
- RE digestion was performed using MluCI ( FIGS. 19E-19G ) and PstI+MslI ( FIGS. 19H-19J ) to eliminate Ex 16/17 and IEJ 3/16 target sequences, respectively.
- FIG. 19E and FIG. 19H illustrate jgISH nuclei.
- FIG. 19F and FIG. 19I illustrate graphs of quantification of average foci per nucleus; statistical significance was determined using the unpaired, two-tailed Mann-Whitney test.
- FIG. 19G and FIG. 19J illustrate cumulative frequency distributions represented as the number of foci per nucleus compared using the unpaired, two-tailed non-parametric Kolmogorov-Smirnov test. ****p ⁇ 0.0001. n.s., not-significant. Error bars are ⁇ SEM. Scale bars are 10 ⁇ m.
- FIG. 20A illustrates a schema of non-classical gencDNAs variants of APP identified by SMRT sequencing from SAD brains.
- FCTX Neuronal nuclei from SAD prefrontal cortex
- FCTX genomic DNA PCR
- Multiple reactions were pooled for (3) library preparation to enable (4) high fidelity sequencing (SMRT 20 ⁇ CCS calling).
- FIG. 20B illustrates intraexonic junctions (IEJs).
- FIG. 20C illustrates a chart of total number and proportion of unique reads from each identified IEJ form.
- FIG. 20D illustrates a graph of number of unique IEJs forms.
- FIG. 20E illustrates a concentric circle plot of the APP locus (exon numbers along perimeter) illustrating IEJs (connecting lines inside the circles), deletions (DEL) (first inner circle), insertions (INS) (second inner circle), and single nucleotide variations (SNVs) (third inner circle) from sporadic Alzheimer's Disease (SAD) brains.
- Black dots indicate the abundance of DELs, INSs, and SNVs on a log(10) scale at the specified exon location.
- the outermost circle illustrates the sum count (key) of unique changes. A ⁇ region is highlighted, and known familial AD mutations are circled.
- FIG. 20F illustrates a concentric circle plot of the APP locus (exon numbers along perimeter) illustrating IEJs (connecting lines inside the circles), deletions (DEL) (first inner circle), insertions (INS) (second inner circle), and single nucleotide variations (SNVs) (third inner circle) from non-diseased brains. Black dots indicate the abundance of DELs, INSs, and SNVs on a log(10) scale at the specified exon location. Outermost circle illustrates the sum count (key) of unique changes. A ⁇ region is highlighted.
- FIG. 21A illustrates 10 different familial AD mutations present in APP gencDNAs.
- In-frame mutations A673V, V715M, I716M, V717F, L723P, and V717I
- out-of-frame mutations V715A, T719P, A713T, T714I are indicated based on the known APP reading frame analysis.
- FIGS. 21B-21C illustrate nuclei sorted from 6 ND and 6 SAD cortices. Nuclei were analyzed by Ex 16/17 ( FIG. 21B ) and IEJ 3/16 jgISH ( FIG. 21C ). Cumulative frequency distribution plots of number of foci per nucleus showed statistical significance (nonparametric Kruskal-Wallis test with Dunn's multiple corrections). ****P ⁇ 0.0001. n.s., not-significant. Error bars are ⁇ SEM. Scale bars are 10 ⁇ m.
- FIGS. 21D-21I illustrate plots of relative percentage of nuclei sorted from 6 ND and 6 SAD cortices. Nuclei were analyzed by Ex 16/17 ( FIGS. 21D-21F ) and IEJ 3/16 ( FIGS. 21G-21I ) jgISH. Cumulative frequency distribution plots of number of foci per nucleus showed statistical significance (nonparametric Kruskal-Wallis test with Dunn's multiple corrections). *p ⁇ 0.05, **p ⁇ 0.01, ***p ⁇ 0.001, ****p ⁇ 0.0001. n.s., not-significant. Error bars are ⁇ SEM.
- FIG. 22A illustrates images of IEJ 3/16 jgISH of nuclei isolated from the cortex of an AD mouse model (J20 transgenic, with neuron specific expression of human APP cDNA containing Swedish and Indiana mutations) versus WT littermates. Error bars are ⁇ SEM. Scale bars are 10 ⁇ m.
- FIGS. 22B-22C illustrate quantification of nuclei from FIG. 22A .
- FIG. 22B illustrates a cumulative frequency distribution illustrating the foci per nucleus.
- FIG. 22C illustrates a graph of an average number of foci per nucleus. ****p ⁇ 0.0001. n.s., not-significant.
- FIGS. 22D-22G illustrate IEJ 3/16 jgISH from two mouse experiments.
- FIG. 22D illustrates a graph of foci per nucleus (x-axis) as relative percent of nuclei (y-axis) from J20+, WT+, J20 ⁇ , and WT ⁇ from a first mouse experiment.
- FIG. 22E illustrates a graph of average foci per nucleus (y-axis) from J20+, WT+, J20 ⁇ , and WT ⁇ mice from a first mouse experiment.
- FIG. 22F illustrates a graph of foci per nucleus (x-axis) as relative percent of nuclei (y-axis) from J20+, WT+, J20 ⁇ , and WT ⁇ from a second mouse experiment.
- FIG. 22G illustrates a graph of average foci per nucleus (y-axis) from J20+, WT+, J20 ⁇ , and WT ⁇ mice from a second mouse experiment.
- FIG. 23A illustrates images of representative nuclei isolated from 177 and 829-day J20 cortices. Scale bars are 10 ⁇ m.
- FIG. 23B illustrates a graph of cumulative percentage distribution of foci area (y-axis) as compared to relative foci area (x-axis) of nuclei from FIG. 23A .
- FIG. 23C illustrates a graph of average foci area (y-axis) of nuclei from FIG. 23A .
- FIG. 23D illustrates synthetic DNA targets containing the Ex 16/17 junction sequence introduced by retroviral transduction in NIH-3T3 cells, and the target sequence (provirus) identified by Ex 16/17 jgISH.
- FIGS. 23E-23F illustrates graphs of foci size from concatamer ( ⁇ 2) from FIG. 23D .
- FIG. 23E illustrates a graph of cumulative percentage of foci (y-axis) as compared to relative foci area (x-axis).
- FIG. 23F illustrates average foci area (x-axis) from x0, x1, x2 of FIG. 23D . ****p ⁇ 0.0001. n.s., not-significant. Error bars are ⁇ SEM.
- FIG. 24 illustrates cortical tissue sections from human Alzheimer's disease subjects stained with antibodies recognizing AP peptide (“A ⁇ ”) and APP c-terminal (“c-terminal”). “Merge” images illustrate co-localization of AP peptide with APP c-terminal. “DAPI” images illustrate nuclei staining.
- non-classical variant refers to RNA or DNA molecules comprising intraexonic junctions between exons and/or conventional spliced exon-exon junctions, and RNA or DNA molecules lacking introns between exons.
- genomic cDNA refers to a genomic variant lacking introns.
- the gencDNA comprises intraexonic junctions between exons.
- the gencDNA comprises inverted exons.
- the gencDNA is generated by reverse transcription of a non-classical RNA variant.
- the gencDNA is incorporated into genomic DNA.
- expression refers to a transcriptional or translational product of a gene.
- activity refers to protein biological or chemical function.
- RNA refers to a molecule comprising at least one ribonucleotide residue.
- ribonucleotide is meant a nucleotide with a hydroxyl group at the 2′ position of a beta-D-ribo-furanose moiety.
- RNA includes, but not limited to, mRNA, ribosomal RNA, tRNA, non-protein-coding RNA (npcRNA), non-messenger RNA, functional RNA (fRNA), long non-coding RNA (lncRNA), pre-mRNAs, and primary miRNAs (pri-miRNAs).
- RNA includes, for example, double-stranded (ds) RNAs; single-stranded RNAs; and isolated RNAs such as partially purified RNA, essentially pure RNA, synthetic RNA, recombinantly produced RNA, as well as altered RNA that differ from naturally-occurring RNA by the addition, deletion, substitution and/or alteration of one or more nucleotides.
- alterations can include addition of non-nucleotide material, such as to the end(s) of the siRNA or internally, for example at one or more nucleotides of the RNA.
- Nucleotides in the RNA molecules described herein can also comprise non-standard nucleotides, such as non-naturally occurring nucleotides or chemically synthesized nucleotides or deoxynucleotides. These altered RNAs can be referred to as analogs or analogs of naturally-occurring RNA.
- RNAi refers to an RNA molecule that induces RNA interference (RNAi).
- the RNAi molecule is a dsRNA molecule that will generate a siRNA molecule or miRNA molecule following contact with Dicer (i.e., an RNAi molecule precursor).
- the RNAi molecule is a siRNA duplex, a siRNA sense molecule, a siRNA anti-sense molecule, a miRNA duplex, a miRNA sense molecule, a miRNA anti-sense molecule, and analogues thereof.
- binding fragment means a portion or fragment of an intact antibody molecule, preferably wherein the fragment retains antigen-binding function.
- antibody fragments include Fab, Fab′, F(ab′) 2 , Fd, Fd′ and Fv fragments, diabodies, linear antibodies, single-chain antibody molecules, single-chain binding polypeptides, scFv, bivalent scFv, tetravalent scFv, and bispecific or multispecific antibodies formed from antibody fragments.
- Fab fragments are typically produced by papain digestion of antibodies resulting in the production of two identical antigen-binding fragments, each with a single antigen-binding site and a residual “Fc” fragment. Pepsin treatment yields a F(ab′)2 fragment that has two antigen-combining sites capable of cross-linking antigen.
- An “Fv” is the minimum antibody fragment that contains a complete antigen recognition and binding site. In a two-chain Fv species, this region consists of a dimer of one heavy- and one light-chain variable domain in tight, non-covalent association.
- scFv single-chain Fv
- one heavy- and one light-chain variable domain are covalently linked by a flexible peptide linker such that the light and heavy chains associate in a “dimeric” structure analogous to that in a two-chain Fv species. It is in this configuration that the three CDRs of each variable domain interact to define an antigen-binding site on the surface of the VH-VL dimer.
- the six CDRs confer antigen-binding specificity to the antibody.
- a single variable domain or half of an Fv comprising only three CDRs specific for an antigen
- the Fab fragment also contains the constant domain of the light chain and the first constant domain (C H 1) of the heavy chain.
- Fab fragments differ from Fab′ fragments by the addition of a few residues at the carboxy terminus of the heavy-chain C H 1 domain including one or more cysteines from the antibody hinge region.
- Fab′-SH is the designation herein for Fab′ in which the cysteine residue(s) of the constant domains bear a free thiol group.
- F(ab′) 2 antibody fragments originally were produced as pairs of Fab′ fragments that have hinge cysteines between them. Other chemical couplings of antibody fragments are also suitable.
- monoclonal antibody refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations that are present in minor amounts.
- monoclonal antibodies are made, for example, by the hybridoma method.
- monoclonal antibodies are isolated from phage antibody libraries.
- the antibodies herein include monoclonal, polyclonal, recombinant, chimeric, humanized, bi-specific, grafted, human, and fragments thereof including antibodies altered by any means to be less immunogenic in humans.
- the monoclonal antibodies and fragments herein include “chimeric” antibodies and “humanized” antibodies.
- chimeric antibodies include a portion of the heavy and/or light chain that is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, so long as they exhibit the desired biological activity.
- a chimeric antibody contains variable regions derived from a mouse and constant regions derived from human in which the constant region contains sequences homologous to both human IgG2 and human IgG4.
- “Humanized” forms of non-human (e.g., murine) antibodies or fragments are chimeric immunoglobulins, immunoglobulin chains or fragments thereof (such as Fv, Fab, Fab′, F(ab′) 2 or other antigen-binding subsequences of antibodies) which contain minimal sequence derived from non-human immunoglobulin.
- Humanized antibodies include, grafted antibodies or CDR grafted antibodies wherein part or all of the amino acid sequence of one or more complementarity determining regions (CDRs) derived from a non-human animal antibody is grafted to an appropriate position of a human antibody while maintaining the desired binding specificity and/or affinity of the original non-human antibody.
- CDRs complementarity determining regions
- corresponding non-human residues replace Fv framework residues of the human immunoglobulin.
- humanized antibodies comprise residues that are found neither in the recipient antibody nor in the imported CDR or framework sequences. These modifications are made to further refine and optimize antibody performance.
- the humanized antibody comprises substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin consensus sequence. Numerous methods for “humanizing” antibodies are known in the art.
- AD Alzheimer's disease
- FAD familial Alzheimer's disease
- SAD sporadic AD
- AD neuropathology is characterized by accumulation of amyloid beta protein and neurofibrillary tangles comprising Tau in the Central Nervous System, synaptic loss, and neuronal death. Specifically, accumulation of amyloid beta as amyloid beta protein plaques or soluble amyloid beta oligomers has been implicated in AD progression.
- Amyloid beta protein results from cleavage of amyloid beta precursor protein (APP) by ⁇ -, ⁇ -, and ⁇ -secretases. In some instances, cleavage of APP is amyloidogenic and results in unwanted accumulation of amyloid beta protein.
- APP amyloid beta precursor protein
- Antibody therapies for treating Alzheimer's disease are primarily aimed at targeting unwanted accumulation of amyloid beta protein. As a result, unwanted amyloid beta protein is cleared from the brain. These treatments, however, result in only partial success. Thus, successful treatments for Alzheimer's disease need to account for the genetic complexity of the disease.
- Amyloid precursor protein (APP) gene copy number has been implicated in AD pathogenesis. For example, Down Syndrome (DS) where there are three copies of APP results in neuropathology similar to AD.
- DS Down Syndrome
- increased APP gene copy number has been observed in AD brains.
- the increased gene copy number is a result of genomic rearrangements such as intraexonic rearrangements that generate non-classical variants of APP.
- Non-classical variants as described herein comprise, in some embodiments, intraexonic rearrangements. In some embodiments, the non-classical variants lack introns. In some embodiments, the non-classical variants comprise a portion of a first exon of APP and a portion of a second exon of APP. In some embodiments, the non-classical variants comprise intraexonic junctions and lack introns. In some embodiments, the non-classical variants comprise inverted exons. In some embodiments, the non-classical variants are genomic cDNAs (gencDNAs). In some embodiments, the non-classical variants comprise a single nucleotide variant (SNV).
- SNV single nucleotide variant
- non-classical variants comprise deletion of exons. In some embodiments, non-classical variants comprise insertions. In some embodiments, non-classical variants comprise copy number variation (CNV), L1 repeat elements, SNVs, deletions, insertions, intraexonic junctions, or combinations thereof. In some embodiments, the SNVs are somatic SNVs. In some embodiments, the SNVs are germline SNVs.
- non-classical variants of APP are generated by DNA damages induced DNA repair.
- the non-classical variants of APP are generated by a homologous recombination event.
- the non-classical variants of APP are generated by a non-homologous recombination event.
- a reverse transcriptase is involved in generating the non-classical variants of APP.
- a non-classical variant of APP is generated by activity of a DNA polymerase.
- generation of the non-classical variants of APP comprises a RNA splicing event.
- Generation of a non-classical variant of APP involves transcription.
- generation of the one or more non-classical variant(s) involves reverse transcription.
- the transcription is cell-type specific.
- the non-classical variants are generated by neuron-specific RNA transcription.
- a non-classical a variant of APP is generated by incorporation of RNA intermediates into genomic DNA.
- the RNA intermediates are non-classical RNA variants of APP.
- the RNA intermediates are reversed transcribed and introduced into the genomic DNA.
- the RNA intermediates that are reversed transcribed are gencDNAs.
- incorporation of RNA intermediates into the genomic DNA involves a break in the DNA.
- the break is a single-stranded break.
- the break is a double-stranded break.
- the break is introduced by an enzyme, a chemical, or radiation.
- generation of a non-classical variant of APP involves alteration in a DNA repair pathway.
- DNA repair pathways include, but are not limited to, non-homologous end joining (NHEJ), microhomology-mediated end joining (MMEJ), homologous recombination, mismatch repair, nucleotide excision repair, or DNA strand cross-link repair.
- generation of a non-classical variant of APP involves RNA processing.
- generation of the non-classical variants of APP comprises a RNA splicing event.
- generation of the non-classical variants of APP comprises a RNA alternative splicing event.
- Exemplary alternative splicing events include, but are not limited, intron retention, exon skipping, alternative 5′ splice site, alternative 3′ splice site, and mutually exclusive exons.
- the non-classical variants are coding. In some embodiments, the non-classical variants are non-coding. In some embodiments, the non-classical variants comprise RNA or DNA. In some embodiments, the non-classical variants comprise genomic cDNA (gencDNA).
- the non-classical variants comprise a portion or all of an exon of APP. In some embodiments, the non-classical variants comprise a portion or all of exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof of APP. In some embodiments, the non-classical variants comprise a portion or all of exon 1, 2, 3, 4, 5, 6, 7, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof of APP. In some embodiments, the non-classical variants do not comprise exon 8. In some embodiments, the non-classical variants do not comprise exon 7. In some embodiments, the non-classical variants do not comprise exon 8 and exon 7.
- the non-classical variants of APP comprise a portion or all of exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18 of APP, wherein the portion or all of the exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18 of APP is inverted.
- the non-classical variants comprise nucleotides 1-24 of exon 3 of APP and nucleotides 45-101 of exon 16 of APP. In some embodiments, the non-classical variants comprise nucleotides from more than one exon of APP. In some embodiments, the non-classical variants comprise nucleotides from at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, or 17 exons of APP. In some embodiments, the non-classical variants comprise a portion of at most 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, or 17 exons of APP.
- non-classical variants of APP comprise a portion of exon 1 and a portion of exon 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof.
- non-classical variants of APP comprise at least or about 10 nucleotides (nt), 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt of exon 1 and at least or about 10 nt, 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt of exon 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof.
- non-classical variants of APP comprise a portion of exon 2 and a portion of exon 1, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof.
- non-classical variants of APP comprise at least or about 10 nucleotides (nt), 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt of exon 2 and at least or about 10 nt, 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt of exon 1, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof.
- non-classical variants of APP comprise a portion of exon 3 and a portion of exon 1, 2, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof.
- non-classical variants of APP comprise at least or about 10 nucleotides (nt), 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt of exon 3 and at least or about 10 nt, 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt of exon 1, 2, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof.
- non-classical variants of APP comprise a portion of exon 4 and a portion of exon 1, 2, 3, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof.
- non-classical variants of APP comprise at least or about 10 nucleotides (nt), 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt of exon 4 and at least or about 10 nt, 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt of exon 1, 2, 3, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof.
- non-classical variants of APP comprise a portion of exon 5 and a portion of exon 1, 2, 3, 4, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof.
- non-classical variants of APP comprise at least or about 10 nucleotides (nt), 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt of exon 5 and at least or about 10 nt, 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt of exon 1, 2, 3, 4, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof.
- non-classical variants of APP comprise a portion of exon 6 and a portion of exon 1, 2, 3, 4, 5, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof.
- non-classical variants of APP comprise at least or about 10 nucleotides (nt), 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt of exon 6 and at least or about 10 nt, 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt of exon 1, 2, 3, 4, 5, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof.
- non-classical variants of APP comprise a portion of exon 7 and a portion of exon 1, 2, 3, 4, 5, 6, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof.
- non-classical variants of APP comprise at least or about 10 nucleotides (nt), 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt of exon 7 and at least or about 10 nt, 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt of exon 1, 2, 3, 4, 5, 6, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof.
- non-classical variants of APP comprise a portion of exon 8 and a portion of exon 1, 2, 3, 4, 5, 6, 7, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof.
- non-classical variants of APP comprise at least or about 10 nucleotides (nt), 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt of exon 8 and at least or about 10 nt, 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt of exon 1, 2, 3, 4, 5, 6, 7, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof.
- non-classical variants of APP comprise a portion of exon 9 and a portion of exon 1, 2, 3, 4, 5, 6, 7, 8, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof.
- non-classical variants of APP comprise at least or about 10 nucleotides (nt), 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt of exon 9 and at least or about 10 nt, 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt of exon 1, 2, 3, 4, 5, 6, 7, 8, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof.
- non-classical variants of APP comprise a portion of exon 10 and a portion of exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof.
- non-classical variants of APP comprise at least or about 10 nucleotides (nt), 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt of exon 10 and at least or about 10 nt, 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt of exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof.
- non-classical variants of APP comprise a portion of exon 11 and a portion of exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 13, 14, 15, 16, 17, 18, or combinations thereof.
- non-classical variants of APP comprise at least or about 10 nucleotides (nt), 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt of exon 11 and at least or about 10 nt, 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt of exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 13, 14, 15, 16, 17, 18, or combinations thereof.
- non-classical variants of APP comprise a portion of exon 12 and a portion of exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 13, 14, 15, 16, 17, 18, or combinations thereof.
- non-classical variants of APP comprise at least or about 10 nucleotides (nt), 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt of exon 12 and at least or about 10 nt, 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt of exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 13, 14, 15, 16, 17, 18, or combinations thereof.
- non-classical variants of APP comprise a portion of exon 13 and a portion of exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 14, 15, 16, 17, 18, or combinations thereof.
- non-classical variants of APP comprise at least or about 10 nucleotides (nt), 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt of exon 13 and at least or about 10 nt, 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt of exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 14, 15, 16, 17, 18, or combinations thereof.
- non-classical variants of APP comprise a portion of exon 14 and a portion of exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 15, 16, 17, 18, or combinations thereof.
- non-classical variants of APP comprise at least or about 10 nucleotides (nt), 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt of exon 14 and at least or about 10 nt, 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt of exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 15, 16, 17, 18, or combinations thereof.
- non-classical variants of APP comprise a portion of exon 15 and a portion of exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 16, 17, 18, or combinations thereof.
- non-classical variants of APP comprise at least or about 10 nucleotides (nt), 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt of exon 15 and at least or about 10 nt, 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt of exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 16, 17, 18, or combinations thereof.
- non-classical variants of APP comprise a portion of exon 16 and a portion of exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 17, 18, or combinations thereof.
- non-classical variants of APP comprise at least or about 10 nucleotides (nt), 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt of exon 16 and at least or about 10 nt, 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt of exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 17, 18, or combinations thereof.
- non-classical variants of APP comprise a portion of exon 17 and a portion of exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 18, or combinations thereof.
- non-classical variants of APP comprise at least or about 10 nucleotides (nt), 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt of exon 17 and at least or about 10 nt, 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt of exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 18, or combinations thereof.
- non-classical variants of APP comprise a portion of exon 18 and a portion of exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or combinations thereof.
- non-classical variants of APP comprise at least or about 10 nucleotides (nt), 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt of exon 18 and at least or about 10 nt, 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt of exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or combinations thereof.
- Non-classical transcript variants as described herein comprise portions of at least 2 exons.
- the non-classical variants comprise a portion of exon 1 and a portion of exon 11.
- the non-classical variants comprise a portion of exon 1 and a portion of exon 12.
- the non-classical variants comprise a portion of exon 1 and a portion of exon 14.
- the non-classical variants comprise a portion of exon 1 and a portion of exon 17.
- the non-classical variants comprise a portion of exon 2 and a portion of exon 14.
- the non-classical variants comprise a portion of exon 2 and a portion of exon 16. In some embodiments, the non-classical variants comprise a portion of exon 2 and a portion of exon 17. In some embodiments, the non-classical variants comprise a portion of exon 2 and a portion of exon 18. In some embodiments, the non-classical variants comprise a portion of exon 3 and a portion of exon 9. In some embodiments, the non-classical variants comprise a portion of exon 3 and a portion of exon 14. In some embodiments, the non-classical variants comprise a portion of exon 3 and a portion of exon 16.
- the non-classical variants comprise a portion of exon 3 and a portion of exon 17. In some embodiments, the non-classical variants comprise a portion of exon 3 and a portion of exon 18. In some embodiments, the non-classical variants comprise a portion of exon 5 and a portion of exon 16. In some embodiments, the non-classical variants comprise a portion of exon comprise exon 6 and a portion of exon 12. In some embodiments, the non-classical variants comprise a portion of exon 6 and a portion of exon 16. In some embodiments, the non-classical variants comprise a portion of exon 6 and a portion of exon 17. In some embodiments, the non-classical variants comprise a portion of exon 6 and a portion of exon 18. In some embodiments, the non-classical variants comprise a portion of exon 16 and a portion exon 18.
- non-classical variants of APP comprising portions of at least two exons, wherein the at least two exons are linked by intraexonic junctions.
- the intraexonic junction is between exon 1 and exon 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof.
- the intraexonic junction is between exon 2 and exon 1, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof.
- the intraexonic junction is between exon 3 and exon 1, 2, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof.
- the intraexonic junction is between exon 4 and exon 1, 2, 3, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof. In some embodiments, the intraexonic junction is between exon 5 and exon 1, 2, 3, 4, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof. In some embodiments, the intraexonic junction is between exon 6 and exon 1, 2, 3, 4, 5, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof. In some embodiments, the intraexonic junction is between exon 7 and exon 1, 2, 3, 4, 5, 6, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof.
- the intraexonic junction is between exon 8 and exon 1, 2, 3, 4, 5, 6, 7, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof. In some embodiments, the intraexonic junction is between exon 9 and exon 1, 2, 3, 4, 5, 6, 7, 8, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof. In some embodiments, the intraexonic junction is between exon 10 and exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof. In some embodiments, the intraexonic junction is between exon 11 and exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 13, 14, 15, 16, 17, 18, or combinations thereof.
- the intraexonic junction is between exon 12 and exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 13, 14, 15, 16, 17, 18, or combinations thereof. In some embodiments, the intraexonic junction is between exon 13 and exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 14, 15, 16, 17, 18, or combinations thereof. In some embodiments, the intraexonic junction is between exon 14 and exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 15, 16, 17, 18, or combinations thereof. In some embodiments, the intraexonic junction is between exon 15 and exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 16, 17, 18, or combinations thereof.
- the intraexonic junction is between exon 16 and exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 17, 18, or combinations thereof. In some embodiments, the intraexonic junction is between exon 17 and exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 18, or combinations thereof. In some embodiments, the intraexonic junction is between exon 18 and exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or combinations thereof.
- sequence complementarity exists in non-classical variants of APP in the intraexonic junctions.
- the sequence complementarity is at least or about 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 50, 60, 70, 80, 90, 100, or more than 100 nucleotides.
- the sequence complementarity is in a range of about 2 nucleotides to 20 nucleotides.
- the sequence complementarity is in a range of about 2 nucleotides to about 200 nucleotides, about 4 nucleotides to about 180 nucleotides, about 6 nucleotides to about 160 nucleotides, about 8 nucleotides to about 140 nucleotides, about 10 nucleotides to about 120 nucleotides, about 12 nucleotides to about 100 nucleotides, about 14 nucleotides to about 80 nucleotides, about 16 nucleotides to about 60 nucleotides, or about 20 nucleotides to about 40 nucleotides.
- Non-classical variants of APP as described herein comprise different sequences.
- a number of different sequences is at least or about 2, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, or more than 1000 sequences.
- non-classical variants of APP comprising portions of at least 2 exons further comprise a deletion of at least one exon or a portion of at least one exon of APP.
- the non-classical transcript variants comprise a deletion of exon 8 or a portion of exon 8 of APP.
- the non-classical transcript variants comprise a deletion of exon 7 or a portion of exon 7 of APP.
- the non-classical transcript variants comprise a deletion of exon or a portion of exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof of APP.
- the non-classical transcript variants comprise a deletion of exon or a portion of exon 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or combinations thereof.
- the APP mRNA sequence is set forth at NCBI Reference Sequence: NM_000484.3. Exemplary non-classical variants are illustrated in Table 1.
- the non-classical variant of APP is a RNA sequence encoding for amyloid beta precursor protein or a portion thereof. In some embodiments, the non-classical variant of APP is a RNA sequence comprising intraexonic junctions between exons of APP. In some embodiments, the non-classical variant of APP is a RNA sequence lacking introns between exons of APP. In some embodiments, the non-classical variant of APP is a RNA sequence comprising one or more single nucleotide variant(s) of APP. In some embodiments, the RNA sequence is set forth as in Table 1.
- the non-classical variant of APP is a DNA sequence encoding for the amyloid beta precursor protein or a portion thereof. In some embodiments, the non-classical variant of APP is a DNA sequence comprising intraexonic junctions between exons of APP or conventional spliced exon-exon junctions present in APP RNA. In some embodiments, the non-classical variant of APP is a DNA sequence lacking introns between exons of APP. In some embodiments, the non-classical variant of APP is a DNA sequence comprising one or more single nucleotide variant(s) of APP. In some embodiments, the non-classical variant of APP is a peptide sequence of a protein encoded by a whole or a part of the non-classical variant of APP.
- non-classical variants of APP wherein the non-classical variants comprise a single nucleotide variation (SNV).
- the non-classical variants comprise one or more SNVs.
- the SNV are familial mutations that arise somatically.
- the SNV are associated with pathogenic Alzheimer's disease.
- Exemplary familial mutations associated with pathogenic Alzheimer's disease which translate to amino acid positions in APP include, but are not limited to, K670, M671, A673, D678, E682, K687, A692, E693, D694, A713, T714, V715, I716, V717, T719, M722, L723, and K724, wherein the amino acids correspond to positions 670, 671, 673, 678, 682, 687, 692, 693, 694, 713, 714, 715, 716, 717, 719, 722, 723, and 724 of SEQ ID NO: 17.
- the SNV in APP which translate to amino acid positions in APP include, but are not limited to, K670N, M671L, A673V, D678H, D678N, E682K, K687N, A692G, E693G, D694N, A713T, T714A, T714I, V715A, V715M, I716F, I716M, I716T, I716V, V717F, V717G, V717I, V717L, T719P, M722K, L723P, and K724N.
- the non-classical variants comprise a SNV in at least one exon of APP. In some embodiments, the non-classical variants comprise one or more SNVs in at least one exon of APP. In some embodiments, the non-classical variants comprise the SNV in one or more exons of APP. In some embodiments, the non-classical variants comprise the SNV in exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof of APP. In some embodiments, the non-classical variants comprise the SNV in exon 17. In some embodiments, the non-classical variants comprise the SNV in the amyloid beta region of APP.
- the SNV in APP which translates to amino acid positions in APP includes, but are not limited to, K670, M671, A673, D678, E682, K687, A692, E693, D694, A713, T714, V715, I716, V717, T719, M722, L723, and K724, wherein the amino acids correspond to positions 670, 671, 673, 678, 682, 687, 692, 693, 694, 713, 714, 715, 716, 717, 719, 722, 723, and 724 of SEQ ID NO: 17.
- the SNV in APP which translates to amino acid positions in APP includes, but are not limited to, A673, A713, T714, V715, I716, V717, T719, and L723, wherein the amino acids correspond to positions 673, 713, 714, 715, 716, 717, 719, and 723 of SEQ ID NO: 17.
- the SNV in APP which translates to amino acid positions in APP includes, but are not limited to, K670N, M671L, A673V, D678H, D678N, E682K, K687N, A692G, E693G, D694N, A713T, T714A, T714I, V715A, V715M, I716F, I716M, I716T, I716V, V717F, V717G, V717I, V717L, T719P, M722K, L723P, and K724N.
- the SNV in APP which translates to amino acid positions in APP includes, but are not limited to, A673V, A713T, T714I, V715M, V715A, I716M, V717I, V717F, T719P, and L723P.
- the non-classical variant of APP comprises a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16. In some embodiments, the non-classical variant of APP comprises at least or about 70%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID NO: 1. In some embodiments, the non-classical variant of APP comprises at least or about 70%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID NO: 2.
- the non-classical variant of APP comprises at least or about 70%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID NO: 3. In some embodiments, the non-classical variant of APP comprises at least or about 70%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID NO: 4.
- the non-classical variant of APP comprises at least or about 70%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID NO: 5. In some embodiments, the non-classical variant of APP comprises at least or about 70%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID NO: 6.
- the non-classical variant of APP comprises at least or about 70%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID NO: 7. In some embodiments, the non-classical variant of APP comprises at least or about 70%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID NO: 8.
- the non-classical variant of APP comprises at least or about 70%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID NO: 9. In some embodiments, the non-classical variant of APP comprises at least or about 70%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID NO: 10.
- the non-classical variant of APP comprises at least or about 70%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID NO: 11. In some embodiments, the non-classical variant of APP comprises at least or about 70%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID NO: 12.
- the non-classical variant of APP comprises at least or about 70%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID NO: 13. In some embodiments, the non-classical variant of APP comprises at least or about 70%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID NO: 14.
- the non-classical variant of APP comprises at least or about 70%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID NO: 15. In some embodiments, the non-classical variant of APP comprises at least or about 70%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID NO: 16.
- a non-classical variant of APP is identified from an individual with Alzheimer's disease (AD).
- a non-classical variant of APP is identified from a brain of an individual with AD.
- a non-classical variant of APP is identified in a neuron isolated from an individual with AD.
- the neuron is sorted and isolated.
- the neuron is sorted and isolated using fluorescence activated nuclear sorting (FANS).
- FANS fluorescence activated nuclear sorting
- a non-classical variant of APP is identified from RNA extracted from a neuron of an individual with AD.
- the RNA is reverse transcribed to cDNA, and a non-classical variant of APP is identified.
- a non-classical variant of APP is identified from genomic DNA extracted from a neuron of an individual with AD.
- AD is sporadic AD.
- AD is familial AD.
- a non-classical variant is identified using a control.
- the control is an individual that does not have AD.
- a non-classical variant of APP is identified using probes that hybridize to full length APP. In some embodiments, a non-classical variant of APP is identified using probes that hybridize to full length APP comprising only exons of APP. In some embodiments, the probes hybridize to genomic DNA. In some embodiments, the probes are used for in situ hybridization. In some embodiments, the probes hybridize to cDNA of APP. An exemplary probe comprises a sequence as set forth in SEQ ID NO: 21. In some embodiments, the probes hybridize to cDNA of APP and are detected by Southern blot. In some embodiments, the Southern Blot is compared to a gel electrophoresis of cDNA of APP. In some embodiments, a non-classical variant of APP is identified by sequencing cDNA corresponding to cDNA detected by the Southern Blot.
- APP amyloid precursor protein
- the biological sample is a blood sample.
- the biological sample comprises RNA, DNA, or protein.
- the disease or disorder is Alzheimer's disease.
- the Alzheimer's disease is familial Alzheimer's disease (FAD) or sporadic Alzheimer's disease.
- the amyloid beta protein is an amyloidogenic protein or a protein that produces an amyloid like morphology. In some embodiments, the amyloid beta protein is encoded by the one or more non-classical variant(s) of APP. In some embodiments, the amyloid beta protein is a variant peptide or protein comprising amyloid beta that is encoded by the one or more non-classical variant(s) of APP. In some embodiments, the amyloid beta protein is a variant peptide or protein that does not comprise amyloid beta and is encoded by the one or more non-classical variant(s) of APP.
- the expression profile is expression level of the one or more non-classical variant(s) of APP. In some embodiments, the expression profile is expression of a set of different non-classical variant of APP. In some embodiments, the activity profile is activity level of the one or more non-classical variant(s) of APP. In some embodiments, the activity profile is activity of a set of different non-classical variant of APP. In some embodiments, the non-classical variant of APP comprises a portion or all of an exon of the APP gene. In some embodiments, the non-classical variant of APP comprises a portion or all of exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof of the APP gene.
- the non-classical variants comprise a portion or all of exon 1, 2, 3, 4, 5, 6, 7, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof of APP. In some embodiments, the non-classical variants do no comprise exon 8. In some embodiments, the one or more exon(s) in the one or more non-classical variant(s) of APP is rearranged as compared to control. In some embodiments, the non-classical variant of APP does not comprise exon 8 of the APP gene. In some embodiments, the non-classical variant of APP comprises a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16.
- the non-classical variant of APP comprises a single nucleotide variation (SNV). In some embodiments, the non-classical variant of APP comprises one or more SNVs. In some embodiments, the SNV in APP, which translates to amino acid positions in APP includes, but are not limited to, K670, M671, A673, D678, E682, K687, A692, E693, D694, A713, T714, V715, I716, V717, T719, M722, L723, and K724, wherein the amino acids correspond to positions 670, 671, 673, 678, 682, 687, 692, 693, 694, 713, 714, 715, 716, 717, 719, 722, 723, and 724 of SEQ ID NO: 17.
- the SNV in APP which translates to amino acid positions in APP includes, but are not limited to, A673, A713, T714, V715, I716, V717, T719, and L723, wherein the amino acids correspond to positions 673, 713, 714, 715, 716, 717, 719, and 723 of SEQ ID NO: 17.
- the SNV in APP which translates to amino acid positions in APP includes, but are not limited to, K670N, M671L, A673V, D678H, D678N, E682K, K687N, A692G, E693G, D694N, A713T, T714A, T714I, V715A, V715M, I716F, I716M, I716T, I716V, V717F, V717G, V717I, V717L, T719P, M722K, L723P, and K724N.
- the SNV in APP which translates to amino acid positions in APP includes, but are not limited to, A673V, A713T, T714I, V715M, V715A, I716M, V717I, V717F, T719P, and L723P.
- the expression profile of one or more non-classical variant(s) of amyloid precursor protein (APP) gene is measured by sequencing of a biological sample from the individual.
- the biological sample comprises RNA or DNA.
- sequencing is performed with any appropriate sequencing technology, including but not limited to single molecule real-time sequencing, Polony sequencing, sequencing by ligation, reversible terminator sequencing, proton detection sequencing, ion semiconductor sequencing, nanopore sequencing, electronic sequencing, pyrosequencing, Maxam-Gilbert sequencing, chain termination (e.g., Sanger) sequencing, +S sequencing, or sequencing by synthesis.
- the expression profile of one or more non-classical variant(s) of APP is measured by nucleotide sequencing.
- the nucleotide sequencing comprises long-read sequencing.
- the long-read sequencing is single molecule real-time sequencing.
- the single molecule real-time sequencing comprises attaching a different fluorescent dye to each of the nucleic acid bases and using a polymerase.
- the polymerase incorporates a single nucleotide comprising the fluorescent dye.
- the fluorescent dye is detected to generate a nucleotide sequence.
- the polymerase is a DNA polymerase or a RNA polymerase.
- the DNA polymerase is a modified polymerase.
- a template to be sequenced is a DNA template or a RNA template.
- the long-read sequencing does not require a reference genome.
- methods for measuring the expression profile comprise preparation of a biological sample prior to sequencing.
- DNA is extracted and purified from the biological sample.
- RNA is extracted.
- RNA is extracted, purified, and reverse transcribed to cDNA.
- the reverse transcribed cDNA or DNA is amplified prior to sequencing.
- single molecule real-time sequencing comprises additional preparation of the biological sample prior to sequencing.
- the DNA is fragmented.
- target regions are amplified to obtain fragmented DNA.
- target regions are enriched by hybridization based DNA pull-down.
- the ends of the DNA are repaired.
- hairpin adapters are ligated to the DNA that then hybridizes to a primer.
- a nuclease is used to remove DNA that did not ligate to the hairpin adapters.
- a DNA polymerase is mixed and the DNA is sequenced.
- the expression profile of one or more non-classical variant(s) of APP is the expression level of one or more non-classical variant(s) of APP.
- the expression level is of a set of different non-classical variants of APP.
- the expression level is measured following long-read sequencing.
- the long-read sequencing is RNA sequencing.
- the long-read sequencing is DNA sequencing.
- the long-read sequencing is single molecule real-time sequencing. During a long-read sequencing reaction, sequenced base pairs or “reads” are generated.
- the expression level is then quantified by counting a number of reads that map to the one or more non-classical variant(s) of APP sequences during the long-read sequencing reaction.
- the one or more non-classical variant(s) of APP sequences comprise a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16.
- the long read sequencing measures a change in the APP gene.
- the change is a change in DNA of the APP gene.
- the DNA is coding DNA.
- the DNA is non-coding DNA.
- the change is a change in RNA of the APP gene.
- the RNA is coding RNA.
- the RNA is non-coding RNA.
- the change is a change in a protein encoded by the APP gene.
- the long read sequencing measures a single nucleotide variation (SNV) in the APP gene. In some embodiments, the long read sequencing measures one or more SNVs in the APP gene. In some embodiments, the SNV in APP, which translates to amino acid positions in APP includes, but are not limited to, K670, M671, A673, D678, E682, K687, A692, E693, D694, A713, T714, V715, I716, V717, T719, M722, L723, and K724, wherein the amino acids correspond to positions 670, 671, 673, 678, 682, 687, 692, 693, 694, 713, 714, 715, 716, 717, 719, 722, 723, and 724 of SEQ ID NO: 17.
- the SNV in APP which translates to amino acid positions in APP includes, but are not limited to, A673, A713, T714, V715, I716, V717, T719, and L723, wherein the amino acids correspond to positions 673, 713, 714, 715, 716, 717, 719, and 723 of SEQ ID NO: 17.
- the SNV in APP which translates to amino acid positions in APP includes, but are not limited to, K670N, M671L, A673V, D678H, D678N, E682K, K687N, A692G, E693G, D694N, A713T, T714A, T714I, V715A, V715M, I716F, I716M, I716T, I716V, V717F, V717G, V717I, V717L, T719P, M722K, L723P, and K724N.
- the SNV in APP which translates to amino acid positions in APP includes, but are not limited to, A673V, A713T, T714I, V715M, V715A, I716M, V717I, V717F, T719P, and L723P.
- the expression profile of one or more non-classical variant(s) of the amyloid precursor protein (APP) gene is measured by a pull-down assay.
- one or more probe(s) for use in the pull-down assay is designed to hybridize to a portion or all of exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof.
- one or more probe(s) for use in the pull-down assay is designed to hybridize to a portion or all of exon 1, 2, 3, 4, 5, 6, 7, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof.
- the one or more probe(s) for use in the pull-down assay is designed to hybridize to a portion or all of a non-classical variant comprising a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16.
- the one or more probe(s) is labeled with an affinity tag.
- affinity tags include, but are not limited to, biotin, desthiobiotin, histidine, polyhistidine, myc, hemagglutinin (HA), FLAG, glutathione S transferase (GST), or derivatives thereof.
- the affinity tag is recognized by avidin, streptavidin, nickel, or glutathione.
- one or more non-classical variant(s) of APP are amplified.
- the one or more non-classical variant(s) of APP are amplified using primers designed to detect exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18.
- the one or more non-classical variant(s) of APP are amplified using primers designed to detect exon 1, 2, 3, 4, 5, 6, 7, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18.
- the primers are used to detect one or more non-classical variant comprising a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16.
- the one or more non-classical variant is amplified by PCR.
- the PCR is qPCR.
- the one or more non-classical variant(s) of APP are not amplified.
- the one or more non-classical variant(s) of APP are visualized using a fluorescent assay, a radioactivity assay, or a luminescent assay.
- the one or more probes used to hybridize to the one or more non-classical variant(s) of APP further comprises a fluorescent tag that is detected using the fluorescent assay.
- the one or more non-classical variant(s) of APP are visualized by gel electrophoresis.
- the pull down assay measures a change in the APP gene.
- the change is a change in DNA of the APP gene.
- the DNA is coding DNA.
- the DNA is non-coding DNA.
- the change is a change in RNA of the APP gene.
- the RNA is coding RNA.
- the RNA is non-coding RNA.
- the change is a change in a protein encoded by the APP gene.
- the expression profile of one or more non-classical variant(s) of the amyloid precursor protein (APP) gene is measured by binding of one or probe(s) to one or more non-classical variant(s) of APP.
- the one or more probe(s) is a polypeptide.
- the one or more probe(s) is a polynucleotide.
- intraexonic rearrangements are detected by measuring binding of the one or more probe(s).
- the one or more probe(s) hybridizes to target sequences within at least two exons of APP gene.
- the one or more probe(s) hybridizes to sequences with one end complementary to a 3′ end of one exon of APP and a second end complementary to a 5′ end of a downstream exon of APP.
- the one or more probe(s) hybridizes to target sequences within at least two exons that are consecutive exons of APP.
- the one or more probe(s) hybridizes to target sequences within at least two exons that are non-consecutive exons of APP.
- the one or more probe(s) are provided in an array.
- the array comprises one or more probe(s) for measuring an expression profile for one or more non-classical variant(s) of APP.
- the one or more probe(s) detects RNA.
- the one or more probe(s) detect exonic rearrangements such as intraexonic rearrangements of APP.
- the one or more probes hybridize to a portion or all of an exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof of APP.
- the one or more probes hybridize to a portion or all of an exon 1, 2, 3, 4, 5, 6, 7, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof of APP.
- the array comprises RNA probes designed to hybridize to the one or more non-classical variant(s) of APP.
- the one or more probe(s) bind to RNA or DNA from the one or more non-classical variant(s) of APP gene. In some embodiments, the one or more probe(s) are used for an amplification reaction. In some embodiments, the amplification reaction is PCR. In some embodiments, the amplification reaction is quantitative such as qPCR. In some embodiments, the PCR reaction utilizes a TaqManTM or a similar quantitative PCR technology. In some embodiments, at least one primer used in the PCR reaction comprises a sequence as set forth in SEQ ID NO: 19 or 20.
- Exemplary methods for detecting binding of the one or more probe(s) include, but are not limited to, enzyme linked immunosorbent assays (ELISA), Western blots, spectroscopy, mass spectrometry, peptide arrays, colorimetry, electrophoresis, isoelectric focusing, immunoprecipitations, immunoassays, and immunofluorescence.
- the expression profile of the one or more non-classical variant(s) of the APP gene is measured by in situ hybridization or immunological hybridization.
- the in situ hybridization is performed without amplification.
- the in situ hybridization is performed without polymerase dependent amplification.
- the in situ hybridization comprises capturing the one or more non-classical variant(s) of APP from the biological sample on a solid support prior to contacting the one or more non-classical variant(s) of APP with the one or more probe(s).
- the in situ hybridization is chromogenic in situ hybridization.
- the in situ hybridization is fluorescence in situ hybridization.
- in situ hybridization allows for detection of intraexonic rearrangements. In some embodiments, in situ hybridization allows for detection for genomic rearrangements such as between introns and exons or between exons and exons.
- the one or more probe(s) for in situ hybridization hybridizes to a region spanning an intraexonic rearrangement. For example, for the non-classical variant cAPP-R3/16, the one or more probe(s) hybridize to a region spanning exon 3 and exon 16. In some embodiments, the one or more probe(s) hybridize to a region of a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16.
- one or more probe(s) that hybridize to an intraexonic junction are non-classical variants of APP comprising portions of at least two exons, wherein the at least two exons are linked by intraexonic junctions.
- the one or more probe(s) hybridize to an intraexonic junction between exon 1 and exon 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof.
- the one or more probe(s) hybridize to an intraexonic junction between exon 2 and exon 1, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof.
- the one or more probe(s) hybridize to an intraexonic junction between exon 3 and exon 1, 2, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof. In some embodiments, the one or more probe(s) hybridize to an intraexonic junction between exon 4 and exon 1, 2, 3, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof. In some embodiments, the one or more probe(s) hybridize to an intraexonic junction between exon 5 and exon 1, 2, 3, 4, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof.
- the one or more probe(s) hybridize to an intraexonic junction between exon 6 and exon 1, 2, 3, 4, 5, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof. In some embodiments, the one or more probe(s) hybridize to an intraexonic junction between exon 7 and exon 1, 2, 3, 4, 5, 6, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof. In some embodiments, the one or more probe(s) hybridize to an intraexonic junction between exon 8 and exon 1, 2, 3, 4, 5, 6, 7, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof.
- the one or more probe(s) hybridize to an intraexonic junction between exon 9 and exon 1, 2, 3, 4, 5, 6, 7, 8, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof. In some embodiments, the one or more probe(s) hybridize to an intraexonic junction between exon 10 and exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof. In some embodiments, the one or more probe(s) hybridize to an intraexonic junction between exon 11 and exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 13, 14, 15, 16, 17, 18, or combinations thereof.
- the one or more probe(s) hybridize to an intraexonic junction between exon 12 and exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 13, 14, 15, 16, 17, 18, or combinations thereof. In some embodiments, the one or more probe(s) hybridize to an intraexonic junction between exon 13 and exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 14, 15, 16, 17, 18, or combinations thereof. In some embodiments, the one or more probe(s) hybridize to an intraexonic junction between exon 14 and exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 15, 16, 17, 18, or combinations thereof.
- the one or more probe(s) hybridize to an intraexonic junction between exon 15 and exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 16, 17, 18, or combinations thereof. In some embodiments, the one or more probe(s) hybridize to an intraexonic junction between exon 16 and exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 17, 18, or combinations thereof. In some embodiments, the one or more probe(s) hybridize to an intraexonic junction between exon 17 and exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 18, or combinations thereof.
- the one or more probe(s) hybridize to an intraexonic junction between exon 18 and exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or combinations thereof. In some embodiments, the one or more probe(s) hybridize to an intraexonic junction between exon 1 and exon 11, exon 1 and exon 14, exon 2 and exon 17, exon 2 and exon 14, exon 2 and exon 18, exon 2 and exon 16, exon 3 and exon 16, exon 3 and exon 14, exon 3 and exon 17, exon 6 and exon 17, exon 6 and exon 18, exon 3 and exon 9, exon 2 and exon 9, exon 16 and exon 18, exon 6 and exon 12, exon 5 and exon 16, or exon 16 and exon 17.
- In situ hybridization comprises probes for detecting one or more non-classical variant(s) of APP.
- the probes hybridize to RNA comprising the one or more non-classical variant(s) of APP.
- the probes hybridize to DNA comprising the one or more non-classical variant(s) of APP.
- the probes hybridize to RNA comprising a portion of a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16.
- the probes comprise a base-pairing region complementary to the target nucleic acid, a spacer sequence, and a base-tail sequence.
- two tail sequences form a hybridization site for amplification.
- the target nucleic acid is visualized following amplification.
- the probes hybridize to protein encoded by the non-classical variant of APP.
- the probes are removed prior to visualization.
- the probes are removed enzymatically, chemically, or mechanically. For example, the probes are removed using restriction enzymes.
- a number of probes are used for in situ hybridization.
- in situ hybridization comprises at least or about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100 or more 100 probes.
- in situ hybridization comprises at most 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, or 100 probes.
- a number of probe pairs are used for in situ hybridization.
- the number of probe pairs comprises at least or about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 45, 50, or more than 50 probe pairs.
- in situ hybridization comprises at most 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 45, or 50 probe pairs.
- following hybridization of a probe or a probe pair the probe or probe pair are amplified prior to visualization.
- a probe is labeled.
- a probe is labeled with a radioactive label, a fluorescent label, an enzyme, a chemiluminescent tag, a colorimetric tag, an affinity tag or other labels or tags that are known in the art.
- affinity tags include, but are not limited to, biotin, desthiobiotin, histidine, polyhistidine, myc, hemagglutinin (HA), FLAG, glutathione S transferase (GST), or derivatives thereof.
- the affinity tag is recognized by avidin, streptavidin, nickel, or glutathione.
- the fluorescent label is a fluorophore, a fluorescent protein, a fluorescent peptide, quantum dots, a fluorescent dye, a fluorescent material, or variations or combinations thereof.
- Exemplary fluorophores include, but are not limited to, Alexa-Fluor dyes (e.g., Alexa Fluor® 350, Alexa Fluor® 405, Alexa Fluor® 430, Alexa Fluor® 488, Alexa Fluor® 500, Alexa Fluor® 514, Alexa Fluor® 532, Alexa Fluor® 546, Alexa Fluor® 555, Alexa Fluor® 568, Alexa Fluor® 594, Alexa Fluor® 610, Alexa Fluor® 633, Alexa Fluor® 647, Alexa Fluor® 660, Alexa Fluor® 680, Alexa Fluor® 700, and Alexa Fluor® 750), APC, Cascade Blue, Cascade Yellow and R-phycoerythrin (PE), DyLight 405, DyLight 488, DyLight 550, DyLight 650, DyLight 680, DyLight 755, DyLight 800, FITC, Pacific Blue, PerCP, Rhodamine, and Texas Red, Cy5,
- fluorescent peptides examples include GFP (Green Fluorescent Protein) or derivatives of GFP (e.g., EBFP, EBFP2, Azurite, mKalamal, ECFP, Cerulean, CyPet, YFP, Citrine, Venus, YPet).
- GFP Green Fluorescent Protein
- derivatives of GFP e.g., EBFP, EBFP2, Azurite, mKalamal, ECFP, Cerulean, CyPet, YFP, Citrine, Venus, YPet.
- fluorescent dyes include, but are not limited to, xanthenes (e.g., rhodamines, rhodols and fluoresceins, and their derivatives); bimanes; coumarins and their derivatives (e.g., umbelliferone and aminomethyl coumarins); aromatic amines (e.g., dansyl; squarate dyes); benzofurans; fluorescent cyanines; indocarbocyanines; carbazoles; dicyanomethylene pyranes; polymethine; oxabenzanthrane; xanthene; pyrylium; carbostyl; perylene; acridone; quinacridone; rubrene; anthracene; coronene; phenanthrecene; pyrene; butadiene; stilbene; porphyrin; pthalocyanine; lanthanide metal chelate complexes; rare-earth metal chelate complex
- the fluorescein dye is, but not limited to, 5-carboxyfluorescein, fluorescein-5-isothiocyanate, fluorescein-6-isothiocyanate and 6-carboxyfluorescein.
- the rhodamine dye is, but not limited to, tetramethylrhodamine-6-isothiocyanate, 5-carboxytetramethylrhodamine, 5-carboxy rhodol derivatives, tetramethyl and tetraethyl rhodamine, diphenyldimethyl and diphenyldiethyl rhodamine, dinaphthyl rhodamine, and rhodamine 101 sulfonyl chloride (sold under the tradename of TEXAS RED®).
- the cyanine dye is Cy3, Cy3B, Cy3.5, Cy5, Cy5.5, Cy7, IRDYE680,
- Fluorescent labels are detected by any suitable method.
- a fluorescent label is detected by exciting the fluorochrome with the appropriate wavelength of light and detecting the resulting fluorescence, e.g., by microscopy, visual inspection, via photographic film, by the use of electronic detectors such as charge coupled devices (CCDs), or photomultipliers.
- the one or more probe(s) are labeled with the same fluorescent label. In some embodiments, the one or more probe(s) are labeled with different fluorescent labels.
- the expression profile is expression level of the one or more non-classical variant(s) of APP.
- the expression level is of a set of different non-classical variants of APP.
- the expression level is measured using PCR. Examples of PCR techniques include, but are not limited to quantitative PCR (qPCR), single cell PCR, PCR-RFLP, digital PCR (dPCR), droplet digital PCR (ddPCR), single marker qPCR, hot start PCR, and Nested PCR.
- the expression level is measured using qPCR.
- the qPCR comprises use of fluorescent dyes or fluorescent probes.
- the fluorescent dye is an intercalating dye. Examples of intercalating dyes include, but are not limited to, intercalating dyes include SYBR green I, SYBR green II, SYBR gold, ethidium bromide, methylene blue, Pyronin Y, DAPI, acridine orange, Blue View, or phycoerythrin.
- the qPCR comprises use of more than one fluorescent probe. In some embodiments, the use of more than one fluorescent probes allows for multiplexing. For example, different non-classical variants are hybridized to different fluorescent probes and can be detected in a single qPCR reaction.
- the probe is used for visualization of the one or more non-classical variant(s) of APP in an individual.
- the probe is visualized by X-Ray, fluoroscopes, ultrasound, CT-scan, PET scan, magnetic resonance image (MRIs), or electromagnetic field.
- the hybridization assay measures a change in the APP gene.
- the change is a change in DNA of the APP gene.
- the DNA is coding DNA.
- the DNA is non-coding DNA.
- the change is a change in RNA of the APP gene.
- the RNA is coding RNA.
- the RNA is non-coding RNA.
- the change is a change in a protein encoded by the APP gene.
- Described herein, in certain embodiments are methods for diagnosing an individual having or suspected of having Alzheimer's disease by measuring the expression profile or the activity profile of the one or more non-classical variant(s) of amyloid precursor protein (APP) gene.
- the expression profile or the activity profile of the one or more non-classical variant(s) of APP is associated with unwanted accumulation of amyloid beta protein but the individual does not have Alzheimer's disease.
- the expression profile or the activity profile is determined from a biological sample from the individual.
- the biological sample comprises RNA or DNA.
- the RNA is pre-mRNA.
- the RNA is mRNA.
- the DNA is nuclear DNA.
- the DNA is extrachromosomal or extranuclear DNA.
- the DNA is circular DNA.
- the biological sample is from a blood sample.
- the blood sample is taken, for example, from the individual by a blood draw.
- the blood sample is processed by centrifugation such as by density centrifugation.
- the blood sample is treated with a red blood cell lysis agent.
- the blood sample comprises cells from the Central Nervous System (e.g., neurons, astrocytes, or microglia) that are released during break down of the blood brain barrier.
- the biological sample is from cerebrospinal fluid.
- the cerebrospinal fluid comprises cells from the Central Nervous System (e.g., neurons, astrocytes, or microglia) that are released during break down of the blood brain barrier.
- a biological sample in some embodiments, comprises exosomes.
- Exosomes are cell-derived vesicles that are released from many cell types including, but not limited to, dendritic cells (DCs), lymphocytes, platelets, mast cells, epithelial cells, endothelial cells, and neurons.
- DCs dendritic cells
- the exosomes are found in blood.
- the exosomes are found in cerebrospinal fluid.
- the biological sample comprises exosomes from the blood.
- the biological sample comprises exosomes from cerebrospinal fluid.
- nucleic acid is extracted from the biological sample.
- the nucleic acid is DNA.
- the DNA is genomic DNA.
- the DNA is extrachromosomal DNA.
- the DNA is circular DNA.
- the nucleic acid is RNA.
- the nucleic acid in some embodiments, is extracted using any technique that does not interfere with subsequent analysis. For example, the nucleic acid is extracted using alcohol precipitation using ethanol, methanol, or isopropyl alcohol. In some embodiments, the nucleic acid is extracted using phenol, chloroform, or any combination thereof. In some embodiments, the nucleic acid is extracted using cesium chloride.
- the nucleic acid is extracted using sodium, potassium or ammonium acetate or any other salt commonly used to precipitate DNA. In some embodiments, the nucleic acid is extracted using utilizes a column or resin based nucleic acid purification. In some embodiments, after extraction the nucleic acid is stored in water, Tris buffer, or Tris-EDTA buffer before subsequent analysis. For example, storage is less than 8° C., 4° C., ⁇ 20° C., or ⁇ 70° C. In some embodiments, the nucleic acid is stored for 1, 2, 3, 4, 5, 6, or 7 days. In some embodiments, the nucleic acid is stored for 1, 2, 3, or 4 weeks. In some embodiments, the nucleic acid is stored for 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 months.
- methods for evaluating an individual for risk of developing a disease or disorder characterized by unwanted accumulation of amyloid beta protein from a biological sample comprise measuring the expression profile or the activity profile of the one or more non-classical variant(s) of APP.
- the one or more non-classical variant(s) of APP comprise a portion or all of an exon of APP gene.
- the one or more non-classical variant(s) of APP comprise a portion or all of exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof.
- the one or more non-classical variant(s) of APP comprise a portion or all of exon 1, 2, 3, 4, 5, 6, 7, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof. In some embodiments, the one or more non-classical variant(s) of APP does not comprise exon 8. In some embodiments, the one or more non-classical variant(s) of APP comprise a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16.
- the non-classical variants comprise a single nucleotide variation (SNV) in APP, which translate to amino acid positions in APP include, but are not limited to, K670, M671, A673, D678, E682, K687, A692, E693, D694, A713, T714, V715, I716, V717, T719, M722, L723, and K724, wherein the amino acids correspond to positions 670, 671, 673, 678, 682, 687, 692, 693, 694, 713, 714, 715, 716, 717, 719, 722, 723, and 724 of SEQ ID NO: 17.
- SNV single nucleotide variation
- the SNV in APP which translate to amino acid positions in APP include, but are not limited to, A673, A713, T714, V715, I716, V717, T719, and L723, wherein the amino acids correspond to positions 673, 713, 714, 715, 716, 717, 719, and 723 of SEQ ID NO: 17.
- the SNV in APP which translate to amino acid positions in APP include, but are not limited to, K670N, M671L, A673V, D678H, D678N, E682K, K687N, A692G, E693G, D694N, A713T, T714A, T714I, V715A, V715M, I716F, I716M, I716T, I716V, V717F, V717G, V717I, V717L, T719P, M722K, L723P, and K724N.
- the SNV in APP which translate to amino acid positions in APP include, but are not limited to, A673V, A713T, T714I, V715M, V715A, I716M, V717I, V717F, T719P, and L723P.
- the non-classical variants comprise one or more SNVs.
- the expression profile is measured by long-read sequencing. In some embodiments, the long read-sequencing is single molecule real-time sequencing. In some embodiments, the expression profile is measured using probe-based assays. For example, the expression profile is measured by quantitative-PCR, in situ hybridization, or pull down assays.
- detecting one or more non-classical variant(s) of an APP gene in an individual in need thereof comprising: detecting an expression profile or an activity profile of the one or more non-classical variant(s) of the APP gene in a biological sample from the individual by a method comprising long-read sequencing of the biological sample.
- the biological sample is a blood sample.
- the biological sample comprises RNA or DNA.
- the one or more non-classical variant(s) of APP comprise a portion or all of an exon of APP gene.
- the one or more non-classical variant(s) of APP comprise a portion or all of exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof. In some embodiments, the one or more non-classical variant(s) of APP comprise a portion or all of exon 1, 2, 3, 4, 5, 6, 7, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof. In some embodiments, the one or more non-classical variant(s) of APP does not comprise exon 8.
- the non-classical variants comprise a single nucleotide variation (SNV) in APP, which translate to amino acid positions in APP include, but are not limited to, K670, M671, A673, D678, E682, K687, A692, E693, D694, A713, T714, V715, I716, V717, T719, M722, L723, and K724, wherein the amino acids correspond to positions 670, 671, 673, 678, 682, 687, 692, 693, 694, 713, 714, 715, 716, 717, 719, 722, 723, and 724 of SEQ ID NO: 17.
- SNV single nucleotide variation
- the SNV in APP which translate to amino acid positions in APP include, but are not limited to, A673, A713, T714, V715, I716, V717, T719, and L723, wherein the amino acids correspond to positions 673, 713, 714, 715, 716, 717, 719, and 723 of SEQ ID NO: 17.
- the SNV in APP which translate to amino acid positions in APP include, but are not limited to, K670N, M671L, A673V, D678H, D678N, E682K, K687N, A692G, E693G, D694N, A713T, T714A, T714I, V715A, V715M, I716F, I716M, I716T, I716V, V717F, V717G, V717I, V717L, T719P, M722K, L723P, and K724N.
- the SNV in APP which translate to amino acid positions in APP include, but are not limited to, A673V, A713T, T714I, V715M, V715A, I716M, V717I, V717F, T719P, and L723P.
- the one or more non-classical variant(s) of APP comprise a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16.
- detecting one or more non-classical variant(s) of an APP gene in an individual in need thereof comprising: detecting an expression profile or an activity profile of the one or more non-classical variant(s) of the APP gene in the biological sample from the individual by a method comprising binding of one or more probe(s) to the one or more non-classical variant(s) of APP.
- the biological sample is a blood sample.
- the biological sample comprises RNA, DNA, or protein.
- the one or more non-classical variant(s) of APP comprise a portion or all of an exon of APP gene.
- the one or more non-classical variant(s) of APP comprise a portion or all of exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof. In some embodiments, the one or more non-classical variant(s) of APP comprise a portion or all of exon 1, 2, 3, 4, 5, 6, 7, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof. In some embodiments, the one or more non-classical variant(s) of APP does not comprise exon 8.
- the single nucleotide variation (SNV) in APP which translates to amino acid positions in APP includes, but are not limited to, K670, M671, A673, D678, E682, K687, A692, E693, D694, A713, T714, V715, I716, V717, T719, M722, L723, and K724, wherein the amino acids correspond to positions 670, 671, 673, 678, 682, 687, 692, 693, 694, 713, 714, 715, 716, 717, 719, 722, 723, and 724 of SEQ ID NO: 17.
- the SNVs in APP which translate to amino acid positions in APP include, but are not limited to, A673, A713, T714, V715, I716, V717, T719, and L723, wherein the amino acids correspond to positions 673, 713, 714, 715, 716, 717, 719, and 723 of SEQ ID NO: 17.
- the SNVs in APP which translate to amino acid positions in APP include, but are not limited to, K670N, M671L, A673V, D678H, D678N, E682K, K687N, A692G, E693G, D694N, A713T, T714A, T714I, V715A, V715M, I716F, I716M, I716T, I716V, V717F, V717G, V717I, V717L, T719P, M722K, L723P, and K724N.
- the SNVs in APP which translate to amino acid positions in APP include, but are not limited to, A673V, A713T, T714I, V715M, V715A, I716M, V717I, V717F, T719P, and L723P.
- the one or more non-classical variant(s) of APP comprise a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16.
- the expression profile or the activity profile of an individual at risk of developing a disease or disorder characterized by unwanted accumulation of amyloid beta protein is compared to a reference expression profile or activity profile from a cohort of control individuals.
- a presence or absence of one or more non-classical variant(s) of APP is compared to the reference expression profile or activity from the cohort of control individuals.
- the expression profile is expression of a set of different non-classical variant(s) of APP.
- the activity profile is activity of a set of different non-classical variant(s) of APP.
- the non-classical variant of APP comprises a portion or all of an exon of the APP gene.
- the non-classical variant of APP comprises a portion or all of exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof of the APP gene. In some embodiments, the one or more non-classical variant(s) of APP comprise a portion or all of exon 1, 2, 3, 4, 5, 6, 7, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof. In some embodiments, the one or more non-classical variant(s) of APP does not comprise exon 8. In some embodiments, the non-classical variant of APP comprises a single nucleotide variation (SNV). In some embodiments, the non-classical variant of APP comprises one or more SNVs.
- SNV single nucleotide variation
- the SNV in APP which translate to amino acid positions in APP include, but are not limited to, K670, M671, A673, D678, E682, K687, A692, E693, D694, A713, T714, V715, I716, V717, T719, M722, L723, and K724, wherein the amino acids correspond to positions 670, 671, 673, 678, 682, 687, 692, 693, 694, 713, 714, 715, 716, 717, 719, 722, 723, and 724 of SEQ ID NO: 17.
- the SNV in APP which translate to amino acid positions in APP include, but are not limited to, A673, A713, T714, V715, I716, V717, T719, and L723, wherein the amino acids correspond to positions 673, 713, 714, 715, 716, 717, 719, and 723 of SEQ ID NO: 17.
- the SNV in APP which translate to amino acid positions in APP include, but are not limited to, K670N, M671L, A673V, D678H, D678N, E682K, K687N, A692G, E693G, D694N, A713T, T714A, T714I, V715A, V715M, I716F, I716M, I716T, I716V, V717F, V717G, V717I, V717L, T719P, M722K, L723P, and K724N.
- the SNV in APP which translate to amino acid positions in APP include, but are not limited to, A673V, A713T, T714I, V715M, V715A, I716M, V717I, V717F, T719P, and L723P.
- the one or more exon(s) in the one or more non-classical variant(s) of APP is rearranged as compared to control.
- the non-classical variant of APP does not comprise exon 8 of the APP gene.
- the non-classical variant of APP comprises a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16.
- the expression profile is expression level. In some embodiments, the expression level of the one or more non-classical variants is at least or about 50%, 100%, 200%, 300%, 400%, 500%, 600%, 700%, or more than 700% increased or elevated as compared to the expression level from the cohort of control individuals. In some embodiments, the activity profile is activity level. In some embodiments, the activity level of the one or more non-classical variants is at least or about 50%, 100%, 200%, 300%, 400%, 500%, 600%, 700%, or more than 700% increased or elevated as compared to the activity level from the cohort of control individuals. In some embodiments, the expression profile or activity profile is used to detect the presence of one or more non-classical variant(s) of APP.
- the expression profile or the activity profile is used to diagnose an individual.
- the individual has a disease or disorder characterized by unwanted accumulation of amyloid beta protein.
- the individual has or is suspected of having Alzheimer's disease.
- the individual is diagnosed with Alzheimer's disease by measuring the expression profile or the activity profile of the one or more non-classical variant(s) of APP.
- the individual is diagnosed with Alzheimer's disease by measuring the expression profile or the activity profile of the one or more non-classical variant(s) of APP and comparing the expression profile or the activity profile to a reference expression profile or activity profile from a cohort of control individuals.
- the individual is diagnosed with Alzheimer's disease when one or more exon(s) in the one or more non-classical variant(s) of APP is rearranged as compared to a control.
- the expression profile is expression level.
- the individual is diagnosed with Alzheimer's disease when the expression level is at least or about 50%, 100%, 200%, 300%, 400%, 500%, 600%, 700%, or more than 700% increased or elevated as compared to the expression level from the cohort of control individuals.
- the activity profile is activity level.
- the individual is diagnosed with Alzheimer's disease when the activity level is at least or about 50%, 100%, 200%, 300%, 400%, 500%, 600%, 700%, or more than 700% increased or elevated as compared to the activity level from the cohort of control individuals.
- the expression profile or the activity profile is used to more accurately diagnose or treat an individual having a disease or disorder.
- use of the expression profile or the activity profile is at least or about 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or more than 95% more accurate at diagnosing a disease or disorder.
- use of the expression profile or the activity profile is at least or about 1.5 ⁇ , 2 ⁇ , 2.5 ⁇ , 3 ⁇ , 3.5 ⁇ , 4.0 ⁇ , 4.5 ⁇ , 5 ⁇ , 6 ⁇ , 7 ⁇ , 8 ⁇ , 9 ⁇ , 10 ⁇ , or more than 10 ⁇ more accurate at diagnosing a disease or disorder.
- the disease or disorder is Alzheimer's disease.
- methods as described herein for accurately diagnosing or treating Alzheimer's disease are improved as compared to methods comprising neurological tests, mental exams, or brain imaging (e.g. MRI, CT, or PET scans).
- determining whether the individual has or is predisposed to Alzheimer's disease is based on the expression profile or the activity profile from, wherein a likelihood of having or being predisposed to Alzheimer's disease is increased when the expression profile or the activity profile is elevated compared to a reference expression profile or reference activity profile of the one or more non-classical variant(s) derived from a cohort of control individuals.
- Methods as described herein for determining a likelihood of having or being predisposed to Alzheimer's disease are improved as compared to methods comprising neurological tests, mental exams, or brain imaging (e.g. MRI, CT, or PET scans).
- the likelihood of having or being predisposed to Alzheimer's disease is increased by at least or about 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or more than 95% by determining the expression profile or activity profile of the one or more non-classical variant(s).
- the likelihood of having or being predisposed to Alzheimer's disease is increased by at least or about 1.5 ⁇ , 2 ⁇ , 2.5 ⁇ , 3 ⁇ , 3.5 ⁇ , 4.0 ⁇ , 4.5 ⁇ , 5 ⁇ , 6 ⁇ , 7 ⁇ , 8 ⁇ , 9 ⁇ , 10 ⁇ , or more than 10 ⁇ by determining the expression profile or activity profile of the one or more non-classical variant(s).
- the expression profile or the activity profile is used for treating an individual having a disease or disorder. In some embodiments, the expression profile or the activity profile is associated with Alzheimer's disease. In some embodiments, a therapeutic agent is administered based on the expression profile or the activity profile. In some embodiments, the therapeutic agent is optimized based on the expression profile or the activity profile. In some embodiments, the expression profile or the activity profile is measured prior to a treatment, during a treatment, or after a treatment.
- the expression profile or the activity profile is measured at 1 day, 2 days, 3 days, 4 days, 5 days 6 days, 1 week, 2 weeks, 3, weeks, 4 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 1 year, 2 years, or more than 2 years before treatment.
- the expression profile or the activity profile is measured at 1 day, 2 days, 3 days, 4 days, 5 days 6 days, 1 week, 2 weeks, 3, weeks, 4 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 1 year, 2 years, or more than 2 years occurs after treatment.
- agents for treating a disease or disorder characterized by unwanted accumulation of amyloid beta protein are agents for treating a disease or disorder characterized by unwanted accumulation of amyloid beta protein.
- methods of treating a disease or disorder characterized by unwanted accumulation of amyloid beta protein in an individual in need thereof comprising: administering to the individual an agent that inhibits activity of one or more non-classical variant(s) of an amyloid precursor protein (APP) gene.
- APP amyloid precursor protein
- methods of diagnosing and treating a disease or disorder characterized by unwanted accumulation of amyloid beta protein in an individual in need thereof comprising: identifying the individual as having the disease or disorder characterized by unwanted accumulation of amyloid beta protein if binding of one or more probe(s) to one or more non-classical variant(s) of an APP gene is measured; and administering to the individual having the expression profile or the activity profile of the one or more non-classical variant(s) of APP an agent that inhibits activity of the one or more non-classical variant(s) of APP.
- methods of diagnosing and treating a disease or disorder characterized by unwanted accumulation of amyloid beta protein in an individual in need thereof comprising: identifying the individual as having the disease or disorder characterized by unwanted accumulation of amyloid beta protein if an expression profile or an activity profile of one or more non-classical variant(s) of an APP gene is measured by a method comprising long-read sequencing of a biological sample from the individual; and administering to the individual having the expression profile or the activity profile of the one or more non-classical variant(s) of APP an agent that inhibits activity of the one or more non-classical variant(s) of APP.
- the agents inhibit an expression profile of the one or more non-classical variant(s) of APP.
- the expression profile is expression level.
- the expression profile is expression of a set of different non-classical variants.
- the agents inhibit activity of the one or more non-classical variant(s) of APP.
- the one or more non-classical variant(s) of APP comprises a portion or all of exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof. In some embodiments, the one or more non-classical variant(s) of APP comprise a portion or all of exon 1, 2, 3, 4, 5, 6, 7, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof. In some embodiments, the one or more non-classical variant(s) of APP does not comprise exon 8. In some embodiments, the one or more exon(s) in the one or more non-classical variant(s) of APP is rearranged as compared to a control. In some embodiments, the one or more non-classical variant(s) of APP comprises a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16.
- the agents inhibit unwanted accumulation of amyloid beta protein. In some embodiments, the unwanted accumulation of amyloid beta protein is associated with a disease or disorder. In some embodiments, the disease or disorder is Alzheimer's disease. In some embodiments, the Alzheimer's disease is familial Alzheimer's disease or sporadic Alzheimer's disease. In some embodiments, the agents inhibit plaque formation comprising a portion of or all amyloid beta. In some embodiments, the agents inhibit soluble amyloid beta protein. In some embodiments, the unwanted accumulation of amyloid beta protein is associated with one or more non-classical variant(s) of APP.
- the agents target one or more non-classical variant(s) of APP gene or protein thereof.
- exemplary agents include, but are not limited to, an antibody, an antigen binding fragment, a RNA interfering agent (RNAi), a small interfering RNA (siRNA), a short hairpin RNA (shRNA), a microRNA (miRNA), an antisense oligonucleotide (AON), a peptide, a peptidomimetic, a small molecule, or an aptamer.
- RNAi RNA interfering agent
- siRNA small interfering RNA
- shRNA short hairpin RNA
- miRNA microRNA
- AON antisense oligonucleotide
- the agent is an N-methyl-D-aspartate (NMDA) receptor antagonist, or an anti-amyloid beta antibody.
- NMDA N-methyl-D-aspartate
- the cholinesterase inhibitor is selected from the group consisting of Donepezil, Galantamine, and Rivastigmine.
- the NMDA receptor antagonist is memantine.
- the anti-amyloid beta antibody is selected from the group consisting of Bapineuzumab, Solanezumab, Gantenerumab, Crenezumab, BAN2401, Ponezumab, and Aducanumab.
- the N-methyl-D-aspartate (NMDA) receptor antagonist, or an anti-amyloid beta antibody is administered in conjunction with an agent that targets one or more non-classical variant(s) of APP gene or protein thereof.
- the agent is an antibody, an antigen binding fragment, a RNA interfering agent (RNAi), a small interfering RNA (siRNA), a short hairpin RNA (shRNA), a microRNA (miRNA), an antisense oligonucleotide (AON), a peptide, a peptidomimetic, a small molecule, or an aptamer that targets the one or more non-classical variant(s) of APP gene or protein thereof.
- RNAi RNA interfering agent
- siRNA small interfering RNA
- shRNA short hairpin RNA
- miRNA microRNA
- AON antisense oligonucleotide
- the N-methyl-D-aspartate (NMDA) receptor antagonist, or an anti-amyloid beta antibody is administered prior to administration of an agent that targets one or more non-classical variant(s) of APP gene or protein thereof.
- the N-methyl-D-aspartate (NMDA) receptor antagonist, or an anti-amyloid beta antibody is administered up to 1 day, up to 2 days, up to 3 days, up to 5 days, or more than 5 days prior to administration of an agent that targets one or more non-classical variant(s) of APP gene or protein thereof.
- the N-methyl-D-aspartate (NMDA) receptor antagonist, or an anti-amyloid beta antibody is administered singly, or over a time course, such as daily, multiple times weekly, weekly, biweekly, monthly or less frequently prior to administration of an agent that targets one or more non-classical variant(s) of APP gene or protein thereof.
- the N-methyl-D-aspartate (NMDA) receptor antagonist, or an anti-amyloid beta antibody is administered prior to administration of an agent that targets one or more non-classical variant(s) of APP gene or protein thereof.
- the N-methyl-D-aspartate (NMDA) receptor antagonist, or an anti-amyloid beta antibody is administered up to 1 day, up to 2 days, up to 3 days, up to 5 days, or more than 5 days prior to administration of an agent that targets one or more non-classical variant(s) of APP gene or protein thereof.
- the N-methyl-D-aspartate (NMDA) receptor antagonist, or an anti-amyloid beta antibody is administered singly, or over a time course, such as daily, multiple times weekly, weekly, biweekly, monthly or less frequently prior to administration of an agent that targets one or more non-classical variant(s) of APP gene or protein thereof.
- the agent is a small molecule.
- the small molecule is an antagonist of APP.
- the small molecule is an antagonist of one or more non-classical variant(s) of APP.
- the one or more non-classical variant(s) of APP comprises a sequence as set forth in SEQ ID NO: 1, 2, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16.
- the small molecule is an antagonist of protein encoded by the APP gene.
- the agent is an antibody.
- Exemplary antibodies include, but are not limited to, a monoclonal antibody, a polyclonal antibody, a bi-specific antibody, a multispecific antibody, a grafted antibody, a human antibody, a humanized antibody, a synthetic antibody, a chimeric antibody, a camelized antibody, a single-chain Fvs (scFv), a single chain antibody, a Fab fragment, a F(ab′) fragment, disulfide-linked Fvs (sdFv), an intrabody, an anti-idiotypic (anti-Id) antibody, or ab antigen-binding fragments thereof.
- the antibody comprises immunoglobulin molecules and immunologically active fragments of immunoglobulin molecules, e.g., molecules that contain an antigen binding site.
- Immunoglobulin molecules are of any type, class (e.g., IgG, IgE, IgM, IgD, IgA and IgY), or subclass (e.g., IgG1, IgG2, IgG3, IgG4, IgA1 and IgA2).
- the antibody selectively binds to a protein encoded by one or more non-classical variant(s) of APP gene. “Selectively binds” refers to the preference of an antibody to interact with one molecule as compared to another.
- the antibody specifically binds to a protein encoded by one or more non-classical variant(s) of APP gene.
- the specified antibodies or binding molecules bind to a particular polypeptide, protein or epitope yet does not bind in a significant or undesirable amount to other molecules present in a biological sample.
- the specified antibody or binding molecule does not undesirably cross-react with non-target antigens and/or epitopes.
- the antibody binds to a protein encoded by the one or more non-classical variant(s) of APP. In some embodiments, the antibody binds to a protein encoded by the one or more non-classical variant(s) of APP comprising a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16. In some embodiments, the antibody does not bind to wild-type protein.
- agents for inhibiting one or more non-classical variant(s) of APP comprise an antisense RNA that hybridizes to a target RNA and inhibits the activity.
- the antisense RNA stringently hybridizes to the target RNA and inhibits the activity.
- the target RNA is one or more non-classical variant.
- the target RNA is one or more non-classical variant comprising a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16.
- the agent is an antisense RNA molecule.
- antisense RNA molecules include, but are not limited to, RNAi, siRNA, shRNA, or miRNA.
- the antisense RNA is double stranded or single stranded.
- the antisense RNA comprises about 1 to about 50 nucleotides.
- the antisense RNA comprises about 5 to about, about 5 to about 30, about 10 to about 30, about 15 to about 25, or about 20 to about 25 nucleotides.
- the antisense RNA is at least or about 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, or 100% complementary to the target RNA.
- the antisense RNA inhibits activity of APP.
- the antisense RNA is a double-stranded antisense RNA molecule (e.g., siRNA, miRNA, shRNA) that down-regulates expression of APP, wherein one of the strands of the double-stranded antisense RNA molecule comprises a nucleotide sequence that is complementary to a nucleotide sequence of APP RNA encoded by APP or a portion thereof, and wherein the second strand of the double-stranded antisense RNA molecule comprises a nucleotide sequence substantially similar to the nucleotide sequence of APP or RNA encoded by APP or a portion thereof.
- the antisense RNA is a double-stranded antisense RNA molecule that down-regulates expression of APP, wherein each strand of the antisense RNA molecule comprises about 15 to 25, 18 to 24, or 19 to about 23 nucleotides, and wherein each strand comprises at least about 14, 17, or 19 nucleotides that are complementary to the nucleotides of the other strand.
- the antisense RNA is a double-stranded antisense RNA molecule that down-regulates expression of APP, wherein each strand of the antisense RNA molecule comprises about 19 to about 23 nucleotides, and wherein each strand comprises at least about 19 nucleotides that are complementary to the nucleotides of the other strand.
- the RNA interfering activity occurs within a cell. In other embodiments, the RNA interfering activity occurs in a reconstituted in vitro system.
- the antisense RNA is a single-stranded antisense RNA molecule that down-regulates expression of APP, wherein the single-stranded antisense RNA molecule comprises a nucleotide sequence that is complementary to a nucleotide sequence of APP or RNA encoded by APP or a portion thereof.
- antisense RNA is a single-stranded antisense RNA molecule that down-regulates expression of APP, wherein the antisense RNA molecule comprises about 15 to 25, 18 to 24, or 19 to about 23 nucleotides.
- antisense RNA molecule is a single-stranded antisense RNA molecule that down-regulates expression of APP, wherein the antisense RNA molecule comprises about 19 to about 23 nucleotides.
- the RNA interfering activity occurs within a cell. In other embodiments, the RNA interfering activity occurs in a reconstituted in vitro system.
- the antisense RNA molecule is a double-stranded polynucleotide molecule comprising self-complementary sense and antisense regions, wherein the antisense region comprises a nucleotide sequence that is complementary to a nucleotide sequence in a target nucleic acid molecule or a portion thereof and the sense region has a nucleotide sequence corresponding to the target nucleic acid sequence or a portion thereof.
- the antisense RNA molecule is assembled from two separate polynucleotides, where one strand is the sense strand and the other is the antisense strand, wherein the antisense and sense strands are self-complementary (e.g., each strand comprises a nucleotide sequence that is complementary to the nucleotide sequence in the other strand; such as where the antisense strand and sense strand form a duplex or double-stranded structure, for example wherein the double-stranded region is about 19, 20, 21, 22, 23, or more base pairs); the antisense strand comprises a nucleotide sequence that is complementary to a nucleotide sequence in a target nucleic acid molecule or a portion thereof and the sense strand comprises a nucleotide sequence corresponding to the target nucleic acid sequence or a portion thereof.
- each strand comprises a nucleotide sequence that is complementary to the nucleotide sequence in the other strand; such as where the antisense strand
- the antisense RNA molecule is assembled from a single oligonucleotide, where the self-complementary sense and antisense regions of the antisense RNA molecule are linked by means of a nucleic acid based or non-nucleic acid-based linker(s).
- the antisense RNA molecule is a polynucleotide with a duplex, asymmetric duplex, hairpin, or asymmetric hairpin secondary structure, having self-complementary sense and antisense regions, wherein the antisense region comprises a nucleotide sequence that is complementary to a nucleotide sequence in a separate target nucleic acid molecule or a portion thereof and the sense region has a nucleotide sequence corresponding to the target nucleic acid sequence or a portion thereof.
- the antisense RNA molecule is a circular single-stranded polynucleotide having two or more loop structures and a stem comprising self-complementary sense and antisense regions, wherein the antisense region comprises a nucleotide sequence that is complementary to a nucleotide sequence in a target nucleic acid molecule or a portion thereof and the sense region has a nucleotide sequence corresponding to the target nucleic acid sequence or a portion thereof, and wherein the circular polynucleotide is processed either in vivo or in vitro to generate an active antisense RNA molecule capable of mediating RNA interfering activity.
- the antisense RNA molecule also comprises a single-stranded polynucleotide having a nucleotide sequence complementary to a nucleotide sequence in a target nucleic acid molecule or a portion thereof, wherein the single stranded polynucleotide further comprises a terminal phosphate group, such as a 5′-phosphate, or 5′,3′-diphosphate.
- an asymmetric duplex is a linear antisense RNA molecule comprising an antisense region, a loop portion that comprises nucleotides or non-nucleotides, and a sense region that comprises fewer nucleotides than the antisense region to the extent that the sense region has enough complimentary nucleotides to base pair with the antisense region and form a duplex with loop.
- an asymmetric hairpin antisense RNA molecule comprises an antisense region having length sufficient to mediate RNA interfering activity in a cell or in vitro system (e.g., about 19 to about 22 nucleotides) and a loop region comprising about 4 to about 8 nucleotides, and a sense region having about 3 to about 18 nucleotides that are complementary to the antisense region.
- the asymmetric hairpin the antisense RNA molecule also comprises a 5′-terminal phosphate group that is chemically modified.
- the loop portion of the asymmetric hairpin antisense RNA molecule comprises nucleotides, non-nucleotides, linker molecules, or conjugate molecules.
- an asymmetric duplex is an antisense RNA molecule having two separate strands comprising a sense region and an antisense region, wherein the sense region comprises fewer nucleotides than the antisense region to the extent that the sense region has enough complimentary nucleotides to base pair with the antisense region and form a duplex.
- an asymmetric duplex antisense RNA molecule comprises an antisense region having length sufficient to mediate RNA interfering activity in a cell or in vitro system (e.g., about 19 to about 22 nucleotides) and a sense region having about 3 to about 18 nucleotides that are complementary to the antisense region.
- an antisense RNA inhibits activity of a target RNA in a cleavage-dependent process.
- the cleavage-dependent process involves the RNA-induced silencing complex (RISC).
- the antisense RNA e.g., siRNA
- the antisense RNA comprises a passenger strand and guide strand.
- the guide strand pairs with a complementary sequence in a mRNA molecule and induces cleavage by an RNase H endonuclease of the RISC complex.
- the RNase H endonuclease is Argonaute.
- an antisense RNA inhibits activity in a cleavage-independent process.
- the antisense RNA e.g., miRNA
- the antisense RNA comprises nucleotide mismatches with their targets and effect gene silencing through translational repression of the target gene.
- an antisense RNA inhibits the one or more non-classical variant(s) of APP gene. In some embodiments, the antisense RNA inhibits pre-mRNA. In some embodiments, the antisense RNA inhibits mRNA. In some embodiments, the antisense RNA alters various functions of the target RNA. In some embodiments, the antisense RNA alters splicing of the RNA to yield one or more mRNA species. In some embodiments, the antisense RNA alters translation of protein from RNA. In some embodiments, the antisense RNA alters translocation of the RNA to the site of protein translation. In some embodiments, the antisense RNA alters a catalytic activity of the RNA or which is facilitated by the RNA. Alternatively or in combination, the antisense RNA reduces an amount of pre-mRNA.
- the one or more non-classical variant(s) comprise exons that are inverted.
- the one or more non-classical variant(s) of APP comprises one or more exons of APP that are inverted.
- the one or more non-classical variant(s) of APP comprises one or more exons of APP and one or more exons of APP that are inverted.
- agents for inhibiting one or more non-classical variant(s) of APP, wherein the one or more exons of APP are inverted comprise a sense RNA that hybridizes to a target RNA and inhibits the activity.
- the sense RNA stringently hybridizes to the target RNA and inhibits the activity.
- the target RNA is one or more non-classical variants of APP comprising inverted exons of APP.
- the agent is a sense RNA molecule.
- the sense RNA is double stranded or single stranded.
- the sense RNA comprises about 1 to about 50 nucleotides.
- the sense RNA comprises about 5 to about, about 5 to about 30, about 10 to about 30, about 15 to about 25, or about 20 to about 25 nucleotides.
- the sense RNA is at least or about 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, or 100% complementary to the target RNA.
- the target RNA comprises inverted exons of APP.
- a sense RNA inhibits the one or more non-classical variant(s) of APP gene, wherein the one or more non-classical variant(s) comprises inverted exons of the APP gene.
- the sense RNA inhibits pre-mRNA.
- the sense RNA inhibits mRNA.
- the sense RNA alters various functions of the target RNA.
- the sense RNA alters splicing of the RNA to yield one or more mRNA species.
- the sense RNA alters translation of protein from RNA.
- the sense RNA alters translocation of the RNA to the site of protein translation.
- the sense RNA alters a catalytic activity of the RNA or which is facilitated by the RNA. Alternatively or in combination, the sense RNA reduces an amount of pre-mRNA.
- the agent is an antisense oligonucleotide (AON).
- the AON comprises antisense oligonucleotide strands.
- the AON comprises sense oligonucleotide strands.
- the AON comprises antisense oligonucleotide strands and sense oligonucleotide strands.
- the AON restores a reading frame and allow for production of functional APP.
- the AON targets RNA of the one or more non-classical variant(s) of APP gene.
- the RNA is pre-mRNA.
- the RNA is mRNA.
- the AON targets DNA of the one or more non-classical variant(s) of APP gene.
- the DNA is genomic DNA.
- the DNA is nuclear DNA.
- the DNA is extrachromosomal or extranuclear DNA.
- the DNA is circular DNA.
- Antisense oligonucleotides in some embodiments, inhibit the expression of one or more non-classical variant(s) of APP gene. In some embodiments, the AONs inhibit the activity of the one or more non-classical variant(s) of APP gene. In some embodiments, the AONs inhibit the expression or activity of the one or more non-classical variant(s) of APP gene by targeting RNA of the one or more non-classical variant(s) of APP gene for degradation.
- antisense oligonucleotides inhibit a gene of a protein involved in transcription of APP.
- An exemplary protein is a transcription factor, coactivator, corepressor, chromatin modifying enzyme, histone acetyltransferase, histone deacetylase, kinase, or methylase, or any other protein involved in a signal transduction pathway that results in transcription of APP.
- the AONs inhibit the gene of a protein involved in transcription of APP to inhibit generation of the one or more non-classical variants of APP.
- the antisense oligonucleotide (AON) results in an insertion, deletion, duplication, or alteration in an incorrectly processed transcript of the APP gene.
- the incorrectly processed transcript of the APP gene is one or more non-classical variant(s) of the APP gene.
- the one or more non-classical variant(s) comprise a portion or all of an exon of APP.
- the one or more non-classical variant(s) comprise a portion or all of exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof of APP.
- the one or more non-classical variant(s) of APP does not comprise exon 8.
- the AON induces exon skipping or exon inclusion to restore the translational reading frame of the one or more non-classical variant(s) of APP.
- the antisense oligonucleotide induces exon skipping.
- the AON is a short nucleic acid sequence that binds to specific mRNA or pre-mRNA sequences of the APP gene to induce exon skipping.
- the AON is a short nucleic acid sequence that binds to specific DNA sequences of the APP gene to induce exon skipping.
- the AON binds splice sites or exonic enhancers. In some embodiments, binding of the AON to specific mRNA or pre-mRNA sequences generates double-stranded regions.
- formation of double-stranded regions occurs at sites where the spliceosome or proteins associated with the spliceosome would normally bind and causes exons to be skipped. In some embodiments, skipping of exons results in restoration of the transcript reading frame and allows for production of functional APP.
- the antisense oligonucleotide induces exon inclusion.
- the AON binds to at least one of a splice site, a site near a splice site, and a site distant to a splice site.
- the AON binds at site in the RNA to prevent disruption of an exon splice enhancer or intron splice enhancer.
- the AON binds at site in the RNA to prevent creation of an exon splice silencer or intron splice silencer.
- the antisense oligonucleotide comprises natural, synthetic, or artificial nucleotide analogues or bases.
- the AON comprises DNA, RNA, or nucleotide analogues.
- the synthetic or artificial nucleotide analogues or bases comprise modifications at one or more of ribose moiety, phosphate moiety, nucleoside moiety, or a combination thereof.
- the antisense oligonucleotide comprises a nucleobase that is unmodified such as adenine, guanine, cytosine, thymine, and uracil or any synthetic or modified nucleobase.
- modified nucleobases include, without limitation, hypoxanthine, xanthine, 7-methylguanine, 5,6-dihydrouracil, 5-methylcytosine, and 5-hydroxymethoylcytosine.
- the antisense oligonucleotide comprises a backbone that connects components of the AON.
- the backbone comprises a 3′-5′ phosphodiester linkage connecting sugar moieties of the AON.
- Examples of a backbone structure or linkages of the AON include, but are not limited to, phosphorothioate, phosphorodithioate, phosphoroselenoate, phosphorodiselenoate, phosphoroanilothioate, phosphoraniladate, and phosphoramidate.
- the backbone structure of the AON does not comprise phosphorous but comprises peptide bonds, for example in a peptide nucleic acid (PNA), or linking groups including carbamate, amides, and linear and cyclic hydrocarbon groups.
- PNA peptide nucleic acid
- the backbone modification is a phosphorothioate linkage. In some embodiments, the backbone modification is a phosphoramidate linkage.
- the antisense oligonucleotide comprises an unmodified sugar moiety such as ribose or deoxyribose or a modified sugar moiety or sugar analog, including a morpholino ring.
- Non-limiting examples of modified sugar moieties include 2′ substitutions such as 2′-O-methyl (2′-O-Me), 2′-O-methoxyethyl (2′MOE), 2′-O-aminoethyl, 2′F; N3′->P5′ phosphoramidate, 2′dimethylaminooxyethoxy, 2′dimethylaminoethoxyethoxy, 2′-guanidinidium, 2′-O-guanidinium ethyl, carbamate modified sugars, and bicyclic modified sugars.
- the sugar moiety modification is an extra bridge bond, such as in a locked nucleic acid (LNA).
- LNA locked nucleic acid
- the sugar analog contains a morpholino ring, such as phosphorodiamidate morpholino (PMO).
- the sugar moiety comprises a ribofuransyl or 2′deoxyribofuransyl modification.
- the sugar moiety comprises 2′4′-constrained 2′O-methyloxyethyl (cMOE) modifications.
- the sugar moiety comprises cEt 2′, 4′ constrained 2′-O ethyl BNA modifications.
- the sugar moiety comprises tricycloDNA (tcDNA) modifications.
- the sugar moiety comprises ethylene nucleic acid (ENA) modifications.
- the sugar moiety comprises MCE modifications.
- the antisense oligonucleotide (AON) comprises an artificial nucleotide analogue.
- exemplary artificial nucleotide analogues include 2′-O-methyl, 2′-O-methoxyethyl (2′-O-MOE), 2′-O-aminopropyl, 2′-deoxy, T-deoxy-2′-fluoro, 2′-O-aminopropyl (2′-O-AP), 2′-O-dimethylaminoethyl (2′-O-DMAOE), 2′-O-dimethylaminopropyl (2′-O-DMAP), T-O-dimethylaminoethyloxyethyl (2′-O-DMAEOE), or 2′-O-N-methylacetamido (2′-O-NMA) modified, LNA, ENA, PNA, HNA, morpholino, methylphosphonate nucleotides, thiolphosphonate nucle
- the antisense oligonucleotide (AON) comprises a number of nucleobases.
- the number of nucleobases comprises a range of about 8 to 50, 8 to 40, 8 to 35, 8 to 30, 8 to 25, 8 to 20, 8 to 15, 9 to 50, 9 to 40, 9 to 35, 9 to 30, 9 to 25, 9 to 20, 9 to 15, 10 to 50, 10 to 40, 10 to 35, 10 to 30, 10 to 25, 10 to 20, 10 to 15, 11 to 50, 11 to 40, 11 to 35, 11 to 30, 11 to 25, 11 to 20, 11 to 15, 12 to 50, 12 to 40, 12 to 35, 12 to 30, 12 to 25, 12 to 20, 12 to 15, 13 to 50, 13 to 40, 13 to 35, 13 to 30, 13 to 25, 13 to 20, 14 to 50, 14 to 40, 14 to 35, 14 to 30, 14 to 25, 14 to 20, 15 to 50, 15 to 40, 15 to 35, 15 to 30, 15 to 25, 15 to 20, 20 to 50, 20 to 40, 20 to 35, 20 to 30, 20 to 25, 25 to 50
- the sequence of the antisense oligonucleotide (AON) is at least or about 40%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, or 99.5% complementary to a target sequence.
- the target sequence is a sequence of the one or more non-classical variant(s) of APP gene.
- the target sequence is a RNA sequence.
- the target sequence is a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16.
- the target sequence is a DNA sequence.
- the agent inhibits transcription of APP. In some embodiments, the agent inhibits transcription of APP and subsequent incorporation of one or more non-classical variants of APP into the genome. In some embodiments, the agent inhibits a protein in a signal transduction pathway involved in the transcription of APP. In some embodiments, the protein is extracellular. Exemplary extracellular proteins include cell membrane receptors including, but not limited to, G protein-coupled receptors, integrin receptors, Notch receptors, cadherin receptors, receptor tyrosine kinase receptors, chemokine receptors, cytokine receptors, death receptors, T-cell receptors, and any combination thereof.
- the agent targets a signaling molecule that signals through the extracellular proteins.
- signaling molecules include, but are not limited to, hormones, neurotransmitters, cytokines, growth factors, cell adhesion molecules, and vitamins.
- the agent targets a signaling molecule of an extracellular protein receptor to prevent binding of the signaling molecule and the extracellular protein to subsequently inhibit transcription of APP.
- the agent mimics a signaling molecule of an extracellular protein receptor to inhibit signaling and subsequent transcription of APP.
- the agent inhibits transcription of APP by inhibiting an intracellular protein involved in transcription of APP.
- the protein is cytosolic.
- the protein is nuclear.
- the protein modulates transcription of APP.
- the protein is a transcription factor, coactivator, corepressor, chromatin modifying enzyme, histone acetyltransferase, histone deacetylase, kinase, or methylase that modulates transcription of APP.
- Exemplary signal transduction pathways involved in transcription of APP include, but are not limited to, Wnt signal transduction pathway, 5′ adenosine monophosphate-activated protein kinase (AMPK), mechanistic target of rapamycin (mTOR) complexes, the Sirtuin 1 (silent mating-type information regulator 2 homolog 1)/peroxisome proliferator-activated receptor gamma co-activator 1- ⁇ (Sirt1/PGC-1 ⁇ ) axis, and cholinergic receptor signaling.
- the agent edits a nucleic acid of one or more non-classical variant(s) of APP.
- the agent edits DNA.
- the agent edits RNA.
- An exemplary system for nucleic acid editing comprises Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) and a CRISPR-associated (Cas) protein. When expressed or transferred into cells alongside a guide RNA (gRNA), a Cas protein allows for the targeted introduction or deletion of genetic information via a complex with CRISPR sequence of mRNA.
- the gRNA comprises a target sequence region, a protospacer-adjacent motif (PAM) region, and a hairpin region.
- PAM protospacer-adjacent motif
- a gRNA shepherds the Cas enzyme to a specific stretch of nucleic acid.
- the gRNA is a single stranded guide RNA (sgRNA).
- the gRNA is a dual stranded guide RNA (dgRNA).
- Cas then cleaves the nucleic acid to disable or repair a gene.
- the nucleic acid is DNA.
- the nucleic acid is RNA.
- a nucleic acid of one or more non-classical variants of APP using a CRISPR/Cas system.
- the CRISPR/Cas system targets DNA of the one or more non-classical variant(s) of APP.
- CRISPR/Cas system targets RNA of the one or more non-classical variant(s) of APP.
- the one or more non-classical variant(s) comprise a portion or all of an exon of APP.
- the one or more non-classical variant(s) comprise a portion or all of exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof of APP. In some embodiments, the one or more non-classical variant(s) of APP comprise a portion or all of exon 1, 2, 3, 4, 5, 6, 7, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof. In some embodiments, the one or more non-classical variant(s) of APP does not comprise exon 8. In some embodiments, the one or more non-classical variant(s) of APP comprise a single nucleotide variation (SNV) in the APP gene.
- SNV single nucleotide variation
- the non-classical variant of APP comprises one or more SNVs in the APP gene.
- the SNV is in the amyloid beta region of APP.
- the SNV in APP, which translate to amino acid positions in APP include, but are not limited to, K670, M671, A673, D678, E682, K687, A692, E693, D694, A713, T714, V715, I716, V717, T719, M722, L723, and K724, wherein the amino acids correspond to positions 670, 671, 673, 678, 682, 687, 692, 693, 694, 713, 714, 715, 716, 717, 719, 722, 723, and 724 of SEQ ID NO: 17.
- the SNV in APP which translate to amino acid positions in APP include, but are not limited to, A673, A713, T714, V715, I716, V717, T719, and L723, wherein the amino acids correspond to positions 673, 713, 714, 715, 716, 717, 719, and 723 of SEQ ID NO: 17.
- the SNV in APP which translate to amino acid positions in APP include, but are not limited to, K670N, M671L, A673V, D678H, D678N, E682K, K687N, A692G, E693G, D694N, A713T, T714A, T714I, V715A, V715M, I716F, I716M, I716T, I716V, V717F, V717G, V717I, V717L, T719P, M722K, L723P, and K724N.
- the SNV in APP which translate to amino acid positions in APP include, but are not limited to, A673V, A713T, T714I, V715M, V715A, I716M, V717I, V717F, T719P, and L723P.
- a nuclease for use in the CRISPR/Cas system is from a species of, but not limited to, Streptococcus, Campylobacter, Nitratifractor, Staphylococcus, Parvibaculum, Roseburia, Neisseria, Gluconacetobacter, Azospirillum, Sphaerochaeta, Lactobacillus, Eubacterium, Corynebacter, Carnobacterium, Rhodobacter, Listeria, Paludibacter, Clostridium, Lachnospiraceae, Clostridiaridium, Leptotrichia, Francisella, Legionella, Alicyclobacillus, Methanomethyophilus, Porphyromonas, Prevotella, Bacteroidetes, Helcococcus, Letospira, Desulfovibrio, Desulfonatronum, Desulfurococcus, Opitutaceae, Tuberibacillus, Bac
- Exemplary Cas proteins include, but are not limited to, Cpf1, C2c1, C2c2, Cas1, Cas1B, Cas2, Cas3, Cas4, Cas5, Cash, Cas7, Cas8, Cas9 (Csn1 or Csx12), Cas10, Cas13, Csy1, Csy2, Csy3, Cse1, Cse2, Csc1, Csc2, Csa5, Csn2, Csm2, Csm3, Csm4, Csm5, Csm6, Cmr1, Cmr3, Cmr4, Cmr5, Cmr6, Csb1, Csb2, Csb3, Csx17, Csx14, Csx10, Csx16, CsaX, Csx3, Csx1, Csx15, Csf1, Csf2, Csf3, Csf4, homologues thereof, and modified versions thereof.
- the Cas protein targets DNA. In some embodiments, the Cas protein targets RNA. In some embodiments, the Cas protein is Cas9. In some embodiments, the Cas protein is Cas13. Cas proteins include, but are not limited to, wild-type Cas and derivatives, chimeras, or mutants thereof.
- the agent modulates generation of one or more non-classical variant(s) of APP as a result of strand breaks. In some embodiments, the agent modulates generation of one or more non-classical variant(s) of APP as a result of single stranded breaks. In some embodiments, the agent modulates generation of one or more non-classical variant(s) of APP as a result of double stranded breaks (DSBs). In some embodiments, the agent inhibits formation of DSBs.
- the agent inhibits formation of DSBs by inhibiting the cause of DSBs including, but not limited to, V(D)J recombination, class switch recombination, meiosis, ionizing radiation, oxidative free radicals, replication across a nick, and inadvertent enzyme actions.
- DSBs in some embodiments, are repaired by a DNA repair pathway.
- alteration in a DNA repair pathway results in an inability or reduced ability to repair DSBs.
- agents for treating a disease or disorder characterized by unwanted accumulation of amyloid beta protein modulate a DNA repair pathway.
- Exemplary DNA repair pathways include, but are not limited to, non-homologous end joining (NHEJ), microhomology-mediated end joining (MMEJ), homologous recombination, mismatch repair, nucleotide excision repair, or DNA strand cross-link repair.
- NHEJ non-homologous end joining
- MMEJ microhomology-mediated end joining
- homologous recombination mismatch repair
- nucleotide excision repair or DNA strand cross-link repair.
- agents for treating a disease or disorder characterized by unwanted accumulation of amyloid beta protein modulate a DNA DSB repair activity.
- the agent targets a gene involved in a DNA repair pathway.
- genes involved in the DNA repair pathway include, but are not limited to, ATM, ATR, MRN, RAD51, BRCA1/BRCA2, KU70/80, DNA-PKcs, Artemis, Ligase IV, and XRCC4.
- the agent targets a protein involved in the DNA repair pathway.
- Exemplary proteins involved in the DNA repair pathway include, but are not limited to, ATM, ATR, CHK1, RAD51, RAD54, PARP1, ERCC1, DNA-PKcs, and Ligase IV.
- the agent inhibits a protein involved in a DNA repair pathway, wherein the DNA repair pathway is non-homologous end joining (NHEJ).
- NHEJ non-homologous end joining
- the protein is involved in a step of NHEJ.
- the protein is involved in DNA termini recognition, bridging of the DNA ends, DNA end processing, or DNA recognition.
- Exemplary proteins involved in NHEJ include, but are not limited to, DNA-PKcs, KU70/80, Artemis, Ligase IV/XRCC4, Pol ⁇ , or Pol ⁇ .
- the agent is an inhibitor of a protein involved in NHEJ.
- Exemplary inhibitors of DNA-PKcs include, but are not limited to, wortmannin, LY294002, NU7026, NU7441, KU-0060648, MSC2490484A, CC-122, and CC-115.
- Exemplary inhibitors of Ligase IV include, but are not limited to, L189 and SCR7.
- the agent inhibits a protein involved in a DNA repair pathway, wherein the DNA repair pathway is homologous recombination.
- the agent inhibits CHK1, MRE1, RAD51, or RAD54.
- the inhibitor of CHK1 is UCN-01.
- the inhibitor of MRE11 is mirin.
- the inhibitor of RAD51 is RI-1 or RI-2.
- the inhibitor of RAD54 is streptonigrin.
- the agent inhibits a protein involved in one or more DNA repair pathways.
- the inhibitor inhibits a protein involved in homologous recombination and non-homologous end joining (NHEJ).
- NHEJ non-homologous end joining
- Exemplary proteins involved in homologous recombination and NHEJ include, but are not limited to, ATM and ATR.
- the inhibitor of ATM is KU55933.
- the inhibitor of ATR is caffeine, VE-821, or NU6027.
- the agent inhibits a protein involved in a DNA repair pathway, wherein the DNA repair pathway comprises repair of single stranded breaks.
- the agent inhibits PARP1.
- Exemplary PARP1 inhibitors include, but are not limited to, Olaparib (AZD2281), Iniparib (BSI 201), Rucaparib (AG014699), Velparib (ABT-888), Talazoparib (BMN-673), CEP 9722, MK 4827, BMN-673, NU1025, E7016, BGB-290, and 3-aminobenzamide.
- APP amyloid precursor protein
- in vitro methods of screening for a therapeutic agent for treating a disease or disorder characterized by unwanted accumulation of amyloid beta protein comprising: contacting a cell that expresses a non-classical variant of an APP gene with a test agent; detecting inhibition of the activity of the non-classical variant of the APP gene as compared to a control; and identifying the test agent as the therapeutic agent if the test agents inhibits activity of the non-classical variant of an APP gene as compared to the control.
- in vitro methods of screening for a therapeutic agent for treating a disease or disorder characterized by unwanted accumulation of amyloid beta protein comprising: contacting a cell that expresses a non-classical variant of an APP gene with a test agent; detecting binding of the test agent to the non-classical variant of the APP gene; and identifying the test agent as the therapeutic agent if the test agents binds to the non-classical variant of the APP gene.
- the therapeutic agents are screened using various methods known in the art.
- the one or more non-classical variant(s) of APP are expressed (e.g., by transfection or transduction) in a cell or organism, contacted with the therapeutic agents, and assayed for changes in activity.
- the cell is a neuron.
- the therapeutic agents are assayed for binding, specificity, stability, or downstream activity.
- the therapeutic agents are screened for inhibition of expression of the non-classical variant of APP gene.
- expression of the non-classical variant of APP is measured by qPCR or gel electrophoresis.
- inhibition of protein expression of a protein encoded by the non-classical variant of the APP gene is measured.
- Exemplary methods for measuring protein expression include, but are not limited to, Western blot, enzyme-linked immunosorbent assays (ELISA), or chromatography.
- chromatography methods include but are not limited to, high-performance liquid chromatography (HPLC) or liquid chromatography-mass spectrometry (LC/MS).
- the therapeutic agents are screened for inhibition of activity of the non-classical variant of APP.
- the activity of the non-classical variant comprises accumulation of amyloid beta protein.
- the accumulation of amyloid beta protein is measured by a method comprising Western blot, enzyme-linked immunosorbent assays (ELISA), or chromatography.
- the therapeutic agents are screened using an enzymatic activity assay or reporter protein activity assay.
- the one or more non-classical variant(s) are engineered to express a reporter gene.
- Exemplary reporter genes include, but are not limited to, acetohydroxyacid synthase (AHAS), alkaline phosphatase (AP), beta galactosidase (LacZ), beta glucuronidase (GUS), chloramphenicol acetyltransferase (CAT), green fluorescent protein (GFP), red fluorescent protein (RFP), yellow fluorescent protein (YFP), cyan fluorescent protein (CFP), horseradish peroxidase (HRP), luciferase (Luc), nopaline synthase (NOS), octopine synthase (OCS), luciferase, and derivatives thereof.
- AHAS acetohydroxyacid synthase
- AP alkaline phosphatase
- LacZ beta galactosidase
- GUS beta glucuronidase
- CAT chloramphenicol acetyltransferase
- GFP green fluorescent protein
- RFP red fluorescent protein
- YFP
- Methods to determine modulation of a reporter gene include, but are not limited to, fluorometric methods (e.g. fluorescence spectroscopy, Fluorescence Activated Cell Sorting (FACS), fluorescence microscopy), and antibiotic resistance determination.
- fluorometric methods e.g. fluorescence spectroscopy, Fluorescence Activated Cell Sorting (FACS), fluorescence microscopy
- antibiotic resistance determination e.g. antibiotic resistance determination.
- therapeutic agents are screened for reducing activity or expression of one or more non-classical variant(s) of APP or protein thereof.
- the therapeutic agents are identified as therapeutic agents if activity is reduced by at least or about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or more than 90%.
- the therapeutic agents are identified as therapeutic agents if expression is reduced by at least or about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or more than 90%.
- the therapeutic agents are identified as therapeutic agents if accumulation of amyloid beta protein is by at least or about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or more than 90%.
- Therapeutic agents are identified by binding assays.
- binding assays include but are not limited to, radioactive binding assays, fluorescence resonance energy transfer, surface plasmon resonance, enzyme-linked immunosorbent assays (ELISA), kinetic exclusion assays, and crystallography assays.
- the therapeutic agents are used for treating an individual in need thereof. Described herein, in certain embodiments, are methods of treating a disease or disorder characterized by unwanted accumulation of amyloid beta protein in an individual in need thereof, comprising: administering to the individual an agent that inhibits activity of one or more non-classical variant(s) of an APP gene.
- the one or more non-classical variant(s) comprises a portion or all of exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof.
- the one or more non-classical variant(s) comprises a portion or all of exon 1, 2, 3, 4, 5, 6, 7, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof.
- the one or more non-classical variant(s) of APP does not comprise exon 8. In some embodiments, the one or more exon(s) in the one or more non-classical variant(s) of APP is rearranged as compared to a control. In some embodiments, the one or more non-classical variant(s) of APP comprises a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16. In some embodiments, the disease or disorder is Alzheimer's disease. In some embodiments, the Alzheimer's disease is familial Alzheimer's disease (FAD) or sporadic Alzheimer's disease.
- FAD familial Alzheimer's disease
- sporadic Alzheimer's disease sporadic Alzheimer's disease.
- the agent that inhibits the activity of APP is an antibody, an antigen binding fragment, a RNA interfering agent (RNAi), a small interfering RNA (siRNA), a short hairpin RNA (shRNA), a microRNA (miRNA), an antisense oligonucleotide, a peptide, a peptidomimetic, a small molecule, or an aptamer.
- RNAi RNA interfering agent
- siRNA small interfering RNA
- shRNA short hairpin RNA
- miRNA microRNA
- the antibody binds to a protein encoded by the one or more non-classical variant(s) of APP.
- the protein is encoded by the one or more non-classical variant(s) of APP comprising a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16.
- kits for identifying one or more non-classical variant(s) of amyloid precursor protein (APP) gene are provided for detecting a portion or all of exon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof. In some embodiments, kits are provided for detecting a portion or all of exon 1, 2, 3, 4, 5, 6, 7, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or combinations thereof.
- kits are provided for detecting intraexonic junction between exon 1 and exon 11, exon 1 and exon 14, exon 2 and exon 17, exon 2 and exon 14, exon 2 and exon 18, exon 2 and exon 16, exon 3 and exon 16, exon 3 and exon 14, exon 3 and exon 17, exon 6 and exon 17, exon 6 and exon 18, exon 3 and exon 9, exon 2 and exon 9, exon 16 and exon 18, exon 6 and exon 12, exon 5 and exon 16, or exon 16 and exon 17.
- kits are provided for detecting the one or more non-classical variant(s) of APP that comprise a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16.
- kits comprise nucleic acid or polypeptide isolation reagents. In some embodiments, kits comprise one or more probe(s) for hybridization or amplification of a target nucleic acid whose expression profile or activity profile is associated with Alzheimer's disease. In some embodiments, kits include one or more probe(s) for control genes, such as housekeeping genes. In some embodiments, the one or more probe(s) for control genes are used, for example, in ⁇ C t calculations. In some embodiments, a probe of the one or more probe(s) is labeled with an enzyme, a radioactive isotope, or a fluorescent label. In some embodiments, the probe is labeled using an affinity tag.
- affinity tags include, but are not limited to, biotin, desthiobiotin, histidine, polyhistidine, myc, hemagglutinin (HA), FLAG, glutathione S transferase (GST), or derivatives thereof.
- the affinity tag is recognized by avidin, streptavidin, nickel, or glutathione.
- the kit comprises a detecting reagent that binds to the one or more probe(s).
- the detecting reagent comprises a radioactive isotope or a fluorescent label
- kits include a carrier, package, or container that is compartmentalized to receive one or more containers such as vials, tubes, and the like, each of the container(s) including one of the separate elements to be used in a method described herein.
- Suitable containers include, for example, bottles, vials, syringes, and test tubes.
- the containers are formed from a variety of materials such as glass or plastic.
- kits comprise one or more additional containers, each with one or more of various materials (such as reagents, optionally in concentrated form, and/or devices) desirable from a commercial and user standpoint for use of described herein.
- materials include, but not limited to, buffers, primers, enzymes, diluents, filters, carrier, package, container, vial and/or tube labels listing contents and/or instructions for use and package inserts with instructions for use.
- a set of instructions is optionally included.
- a label is on or associated with the container.
- a label is on a container when letters, numbers or other characters forming the label are attached, molded or etched into the container itself; a label is associated with a container when it is present within a receptacle or carrier that also holds the container, e.g., as a package insert.
- a label is used to indicate that the contents are to be used for a specific therapeutic application.
- a label also indicates directions for use of the contents, such as in the methods described herein.
- Non-classical variants were identified from neurons isolated from non-diseased brains and Alzheimer's disease brains.
- Neuronal nuclei were isolated from postmortem frontal cortices (CTX) and cerebellums (CBL) of non-diseased (Non-AD) and Alzheimer's disease (AD) brains and prepared for fluorescence activated cell sorting (FACS). Isolated nuclei were fixed and labeled with rabbit anti-NeuN antibody (1:800) (Millipore, Germany) and Alexa Fluor 488 donkey anti-rabbit IgG secondary (1:500) (Life Technologies, Carlsbad, Calif.), and counterstained with propidium iodide, PI (50 ⁇ g/ml) (Sigma, St. Louis, Mo.). Electronically gated diploid neuronal nuclei, determined by PI fluorescence and immunolabeling, were analyzed and sorted.
- PCR polymerase-chain reactions
- a cDNA library was prepared from populations of 50-nuclei from non-diseased (Non-AD) and Alzheimer's disease (AD) brains with lambda phage library system (Clonetech Laboratories).
- Percentages of amyloid beta positive phage clones from Non-AD and AD brains were also determined using probes for amyloid beta (GATGCAGAATTCCGACATGACTCAGGATATGAAGTTCATCATCAAAAATTGGTG TTCTTTGCAGAAGATGTGGGTTCAAACAAAGGTGCAATCATTGGACTCATGGTGG GCGGTGTTGTCATAGCG) (SEQ ID NO: 22) and APP cDNA ( FIG. 6A ).
- a significant increase in percentage of amyloid beta was seen in AD brains than non-AD brains ( FIG. 6B ).
- FIG. 7 shows that full-length APP mRNA is mainly in the cytoplasm.
- Neuronal nuclei were sorted from human postmortem frontal cortices from non-diseased and Alzheimer's disease brains.
- Genomic DNA was extracted and purified using DNeasy Blood and Tissue Kit (Qiagen, Valencia, Calif.). Purified genomic DNA was used as a template for PCR amplification using primers for APP comprising a forward primer (ATGCTGCCCGGTTTGGCA) (SEQ ID NO: 23) and a reverse primer (CTAGTTCTGCATCTGCTCAAAGAACTTG) (SEQ ID NO: 24).
- PCR products amplified with APP primers were run on an agarose gel ( FIG. 8A ) as well as a no template control (NTC).
- PSEN1 primers were used to amplify the purified genomic DNA and were run on an agarose gel with a positive control (PC) and a no template control (NTC) ( FIG. 8B ). PCR products were cloned and sequenced for variant identification.
- non-classical variants of APP were identified in neuronal and non-neuronal gDNA. Using primers for PSEN1, non-classical variants were not identified on gDNA ( FIG. 8B ).
- Genomic DNA from sorted nuclei was extracted by QuickExtract DNA extraction solution (Epicentre) and preamplified by TaqMan PreAmp Master Mix (Thermo Fisher Scientific). Standard qPCR reactions using TaqMan probe based assays were performed in triplicate. Reactions were run on a BioRad qPCR thermocycler using TaqMan Real-Time PCR Master Mix (Thermo Fisher Scientific). The crossing threshold (Ct) was determined for primers for cAPP-R3/16, PSEN1 and TERT within the linear region of the amplification curve.
- the non-classical variant cAPP-R3/16 was quantitatively detected in neuronal and non-neuronal cells with a wide range of distribution whereas as PSEN1 was with a consistent distribution.
- Non-classical variants were detected using a DNA pull-down assay.
- the in vitro transcribed RNA probe sequence used for DNA pull-down was APP cDNA sequence.
- the pull-down sequences were cloned and sequenced for APP variant analysis.
- Neuronal nuclei were isolated from frontal cortices of AD brains as described in Example 1 and analyzed for intron/exon and exon/exon sequences of APP.
- gDNA in nuclei were hybridized with intron/exon and exon/exon probes and labeled with different colors by chromogenic method. Briefly, neuronal nuclei from AD brains were fixed and sorted for NeuN positivity, and dried onto slides. Neuronal nuclei were treated with RNase cocktail (Ambion) for 1 hour at 40° C., following by hydrogen peroxide treatment for 10 minutes at room temperature and protease treatment for 10 minutes at 40° C. DNA denaturation was performed by incubating the slides with 0.58 ⁇ SSC, 70% formamide, and 0.1% SDS for 20 minutes at 80° C. DNA in situ hybridization probes were incubated with samples at 40° C. overnight.
- RNase cocktail Ambion
- Chromogenic developing procedures were performed according to manufacturer's protocol (Advanced Cell Diagnostics). Slides were then visualized by microscopy.
- genomic rearrangements were observed. Specifically, exon-exon junctions as seen in the arrow were present.
- Brain samples from patients with AD were isolated and sectioned, and non-classical variants were analyzed.
- Non-classical variants were analyzed using RNA in situ hybridization. 10 ⁇ m human AD frontal cortices were sectioned and fixed by neutral buffered formalin. Fixed tissue sections were treated with hydrogen peroxide for 10 minutes at room temperature, followed by target retrieval and protease treatment. RNA in situ hybridization probes were incubated with samples for 2 hours at 40° C. Chromogenic developing procedures were performed according to manufacturer's protocol. After RNA in situ hybridization, anti-A13 monoclonal antibody (MOAB, Millipore) was incubated with sample at room temperature overnight. Horseradish peroxidase based developing method was used for signal detection. Slides were then visualized by microscopy at 200 ⁇ and 630 ⁇ magnification.
- MOAB monoclonal antibody
- Non-classical variants were detected by single molecule real-time (SMRT) sequencing.
- RNA Samples were prepared from 2 AD temporal lobes. Methods for preparation of RNA for sequencing were provided according to manufacturer's instructions (Pacific Biosciences). Briefly, target cDNA was prepared and captured by xGene lockdown probes. cDNA of interest was then ligated to adaptors and ready for SMRT sequencing with RSII sequencer.
- Non-neuronal and neuronal nuclei were isolated from non-diseased and AD brains as described in Example 1 and analyzed for exon-exon junctions and intraexonic rearrangements of APP.
- Non-classical variants were analyzed using DNA in situ hybridization. Briefly, nuclei dried on to slides were treated with RNase cocktail for 1 hour at 40° C., followed by hydrogen peroxidase treatment, target retrieval, protease treatment, and DNA denaturation. DNA in situ hybridization probes were incubated with samples at 40° C. overnight. Chromogenic developing procedures were performed according to manufacturer's protocol. Probes used here were designed to detect exon 16 and exon 17.
- Nuclei from neuronal and non-neuronal cells showed an increased signal in AD brains as compared to non-diseased brains ( FIG. 14 ).
- Non-classical variants were analyzed for ability to express protein.
- Non-classical variants cAPP-R3/16, cAPP-R2/16, and ncAPP-R1/4 were epitope tagged with hemagglutinin (HA).
- the non-classical variants were transfected and expressed in cells. Referring to FIG. 15A , nuclei were stained with DAPI (right panel). The epitope tagged non-classical variants also heterologously expressed EGFP (center panel). Non-classical variants comprising coding regions expressed HA-tagged protein (left panel). Western blot for HA showed similar results in that non-classical variants comprising coding regions expressed HA-tagged protein ( FIG. 15B ).
- LN-229 cells were transfected with vehicle, non-classical variant cAPP-R3/16, or wild-type APP cDNA (APP).
- RNA was extracted from the LN-229 cells and subject to reverse transcription PCR (RT-PCR). RT-PCR products were run on a gel. The non-classical variant cAPP-R3/16 and APP were detected ( FIG. 16A ).
- LN-229 cells were also transfected with vehicle, non-classical variant cAPP-R3/16, or APP for analysis by RNA in situ hybridization.
- Cells were fixed in neutral buffered formalin for 10 min at room temperature. Fixed cells were treated with hydrogen peroxide, following by target retrieval and protease treatment.
- RNA in situ hybridization probes were incubated with samples for 2 hours at 40° C. Chromogenic developing procedures were performed according to manufacturer's protocol. Probes used for staining were cAPP-R3/16 and PPM as a positive control.
- the non-classical variant cAPP-R3/16 was detected in cells expressing non-classical variant cAPP-R3/16 as well as in cells expressing wild-type APP ( FIG. 16B ).
- nuclei were fixed in 1:10 diluted buffered formalin (Fisher Healthcare) for 5 minutes. Fixed or unfixed nuclei were then labeled with anti-NeuN rabbit monoclonal antibody (1:800) (Millipore, Germany) and Alexa Fluor 488 donkey anti-rabbit IgG (1:500) (Life Technology, Carlsbad, Calif.), and counterstained with propidium iodide (PI) (50 ⁇ /ml) (Sigma, St. Louis, Mo.). Diploid NeuN positive and negative nuclei were gated by PI and immunofluorescence, and sorted into appropriate populations for RT-PCR or genomic DNA PCR and in situ hybridization. FANS was performed by FACS-Aria with a FACS-Aria II.
- RNA extraction from 50-nuclei populations and bulk tissues were performed using Quick-RNA MicroPrep (Zymo Research, Irvine, Calif.) and RNAeasy Mini kits (Qiagen, Valencia, Calif.), respectively, according to manufacturer's protocol.
- OneStep Ahead RT-PCR (Qiagen, Valencia, Calif.) was used for RT-PCR with APP sense primer 5′-ATGCTGCCCGGTTTGGCA-3′ (SEQ ID NO: 25) and APP anti-sense primer 5′-CTAGTTCTGCATCTGCTCAAAGAACTTG-3′ (SEQ ID NO: 26).
- Low annealing stringency PCR was carried out with the following thermal cycling steps: 95° C. 15 seconds, 55° C. 15 seconds, and 68° C. 2.5 minutes.
- RT-PCR products were run on an agarose gel, denatured, and transferred to a positively charged nylon membrane. UV crosslinked membranes were incubated with denatured and purified 32 P-labelled APP cDNA probes at 42° C. overnight. Blots were washed four times with increasing washing stringency. Images were developed by Typhoon (GE Healthcare Life Sciences) or Fujifilm FLA-5100 phosphorimager.
- DNA extraction from isolated neuronal nuclei populations was performed using DNAeasy and QIAamp DNA Mini kits (Qiagen, Valencia, Calif.) according to manufacturer's instruction.
- High annealing stringency PCR for APP was performed by FastStart PCR master (Sigma, St. Louis, Mo.) with 95° C. 30 seconds, 65° C. 30 seconds, and 72° C. 2.5 minutes, and Platinum SuperFi DNA polymerase (Life Technology) with 98° C. 10 seconds, 65° C. 10 seconds, and 72° C. 1.5 minutes.
- the primer sequences were the following: sense 5′-ATGACAGAGTTACCTGCACC-3′ (SEQ ID NO: 27) and anti-sense 5′-CTAGATATAAAATTGATGGAA-3′ (SEQ ID NO: 28).
- Thermal cycling steps were 95° C. 30 seconds, 52° C. 30 seconds, 72° C. 2 minutes, and 98° C. 10 seconds, 52° C. 10 seconds, 72° C. 1 minute for FastStart PCR master and Platinum SuperFi DNA polymerase, respectively.
- jgISH Junction-Specific Genomic In situ Hybridization
- RNA-ISH RNA-ISH
- sorted nuclei were dried on Plus Gold slides (Fisher Scientific, Pittsburgh, Pa.). Nuclei were then treated with RNase cocktail enzyme mix (1:50) (ThermoFisher) at 40° C. for 60 minutes, followed by 1:10 dilution buffered formalin fixation at room temperature for 5 minutes. After two washes with distilled water, slides were treated with hydrogen peroxide at room temperature for 10 minutes, target retrieval reagent at 95° C. for 15 minutes, followed by protease treatment at 40° C. for 10 minutes. Restriction enzyme was applied after protease treatment for 2 hours if needed.
- RNase cocktail enzyme mix (1:50) (ThermoFisher) at 40° C. for 60 minutes, followed by 1:10 dilution buffered formalin fixation at room temperature for 5 minutes. After two washes with distilled water, slides were treated with hydrogen peroxide at room temperature for 10 minutes, target retrieval reagent at 95° C. for 15 minutes, followed by protease treatment at 40°
- RNA-ISH pretreatment 10 ⁇ m fresh frozen human tissue sections were fixed by 1:10 dilution buffered formalin on ice for 10 minutes. After two washes with PBS, tissue sections were soaked in serial diluted ethanol (50%, 70% and 100%) for 5 minutes at each step. Slides were then treated with hydrogen peroxide at room temperature for 10 minutes, followed by protease at room temperature for 20 minutes. Probes were incubated with tissue sections at 40° C. for 2 hours.
- Hydrogen peroxide, 10 ⁇ target retrieval buffer, proteases, probes (Ex16/17 targeting ACATGACTCAGGATATGAAGTTCATCATCAAAAATTGGTGT TCTTTGCA (SEQ ID NO: 29); IEJ 3/16 targeting TGCCAAGAAGTCTACCCTGAACTGCAGATCACCAAGATGGATGC (SEQ ID NO: 30, including sense and anti-sense probes) and reagents for signal developing were all purchased from Advanced Cell Diagnosis (ACD, Newark, Calif.). Nuclei or tissue sections were counterstained with hematoxylin. Zeiss AX10 Imager.M2 microscope and ZEN2 software were used for image acquisition. Images were thresholded, and foci number/size were quantified using ImageJ for statistical analysis.
- Neuronal DNA was used as template for APP PCR by Platinum SuperFi DNA polymerase with high annealing stringency (98° C. 10 seconds, 65° C. 10 seconds, and 72° C. 1.5 minutes). Multiple PCR reactions were pooled and purified by DNA Clean and Concentrator-5 (Zymo Research, Irvine, Calif.) for SMRT sequencing library preparation. PCR products were repaired using SMRTbell template prep kit version 2.0 (PacBio) and purified using AMPure PB beads (PacBio). Adapters were ligated to DNA to create SMRTbell libraries. Sequencing polymerase was annealed, and the SMRTbell library was loaded using Magbead binding.
- Raw bam sequencing files were converted to fastq format using the ccs2 algorithm in SMRTLink Version 4.0. Reads were only included in the analyzed fastq file if 1) there were more than 20 passes of the sequencing polymerase over the DNA molecule in the zero mode waveguide well and 2) the read was calculated to possess a >0.9999 predicted accuracy.
- Novel algorithms were developed to detect and analyze exon rearrangement in genes of interest.
- the algorithms were specifically designed to analyze long-read sequences generated by Pacific Biosciences Sequel platform.
- a series of quality control (QC) procedures were performed prior to sequence processing to ensure high quality of reads being analyzed.
- PacBio circular consensus sequences CCS reads with less than 20 passes were filtered out to ensure overall sequence quality. Quality score and read length distributions are examined: for APP gene PCR enriched sequences, average median read-wide Phred score is 93 and read length ranged from 64 to 2470 nucleotides. Reads for which the median Phred score was >85 were analyzed.
- BLAST command line tool “blastn” 2.6.0+
- gap open penalty 0 gap extension penalty 2. Any read where both primers were not detected was filtered out.
- reads on the negative strand were reverse complemented in this step.
- BLAST seed length was optimized to avoid ambiguity and ensure sensitivity.
- Ensembl reference sequence for APP protein was downloaded from the GRCh38 reference human genome assembly using the UCSC Genome Browser (http://genome.ucsc.edu/cgi-bin/hgGateway) with RefSeq accession number NM_000484.3. Since the PCR primers started at the start codon and end with the stop codon, sequences of exons 1 and 18 were trimmed to these positions so only the coding sequence of each of the 18 exons was kept and stored as a FASTA file. BLAST was then used to look for local alignment between 18 exons and each quality-filtered CCS read; blastn parameters used: -outfmt 6, -wordsize 25, -gapopen 0, -gapextend 2. These resulting alignment coordinates were used to mark regions of each read covered by exons for analysis of exon arrangements, lengths and patterns of exon-exon joins.
- Phosphorylated oligonucleotides composed of human APP exon 16 and exon 17 sequences with BamHI and BglII restriction sites on the 5′ ends were annealed, and ligated into the BamHI site of the retroviral expression vector S-003-AB LZRSpBMN-linker-IRES-EGFP. Single and concatamerized oligonucleotide inserts were identified by PCR using primers flanking the BamHI insertion site and identified clones were sequenced to confirm insert copy number (GENEWIZ).
- Helper-free ecotropic virus was produced by transfecting DNA constructs (Lipofecatime 2000, Thermo Fisher Scientific) with single or multiple copies of the oligonucleotide inserts into the retrovirus packaging line Phoenix-ECO. 48 hours post-transfection, retroviral supernatants were harvested and 2 mL of selected virus was used for transduction of NIH-3T3 cells in 6 well plates. Retroviral transduction was carried out by removing the cell growth medium, replacing it with 2 mL of retroviral supernatant containing 4 ⁇ g/ml polybrene, and spinning at 25° C. for 1 hour at 2800 r.p.m.
- NIH-3T3 cells were purchased from ATCC. Cells were maintained in Dulbecco's modified Eagle's medium (Invitrogen) containing 5% fetal bovine serum (Invitrogen) at 37° C. under 5% CO2.
- Invitrogen Dulbecco's modified Eagle's medium
- fetal bovine serum Invitrogen
- Non-classical variants were analyzed in transcriptionally amplified RNA from populations of neuronal nuclei.
- Non-classical variant sequences of APP were analyzed by RT-PCR in nuclei isolated by fluorescence activated nuclear sorting (FANS).
- FANS fluorescence activated nuclear sorting
- the workflow ( FIG. 17A ) commenced with FANS to isolate neurons from both non-diseased and verified SAD prefrontal cerebral cortex (Table 5), which were run in parallel. Groups of 50, NeuN-positive neuronal nuclei were isolated and processed for RT-PCR ( FIG. 17A ).
- Validated primers capable of amplifying full-length APP cDNA (APP 770, NM_000484.3) were used, followed by agarose gel electrophoresis.
- splice variants APP 751 (NM_201413.2) and APP 695 (NM_201414.2) were detected. Smaller bands of varied sizes were also detected ( FIG. 17B ).
- RT-PCR on bulk RNA detected the highly expressed canonical APP 751 splice variants as the major product as well as smaller bands ( FIG. 17C ).
- These RT-PCR products were Southern blotted with 32 P-labeled APP cDNA probes ( FIG. 17D ), which produced positive bands from duplicate gels, that were cloned and Sanger sequenced.
- APP splice variants 751 and 695 as well as non-classical variants of APP were detected and characterized by loss of central exons with proximal and distal exons linked by intraexonic junctions (IEJs) ( FIG. 17E ).
- FIG. 17F Twelve non-classical variant sequences with IEJs were identified ( FIG. 17F ).
- One non-classical variant sequence was characterized by an IEJ between the 24 th nucleotide of exon 3 and 45 th nucleotide of exon 16 ( FIG. 17F , “R3/16”).
- the sequence complementarity of joined exons was found in 11 IEJs ranging in overlap from 2 to 20 nucleotides ( FIG. 17G ).
- PCR artifacts were ruled out using independently produced long-read RNAseq data sets derived from oligo-dT-primed RNA from whole SAD brain and SAD temporal lobe, which yielded non-classical variants with similar IEJs ( FIG. 17H ).
- Non-classical variants of APP were analyzed in genomic DNA from populations of neuronal nuclei.
- FIG. 18A High-stringency amplification using the APP primers described above was pursued on thoroughly RNased DNA obtained from sets of 20 neuronal nuclei from both normal and SAD brains ( FIG. 18A ). PCR of nuclear genomic DNA generated clear bands that were similar in size to non-classical variants from RNA-derived RT-PCR products ( FIG. 18B , ⁇ 100-2,300 bp). Interrogation of a second AD related gene, Presenilin 1 (PSEN1), did not produce products from genomic DNA ( FIG. 18B ; 94 Kb).
- PSEN1 Presenilin 1
- genomic cDNAs showing precise exon::exon junctions, central exon deletions, and IEJs, including some species with sequences identical to the non-classical RNA variants identified ( FIG. 18C ).
- APP gencDNA junctions within single neuronal genomes was analyzed using jgISH.
- RNA-ISH RNA-ISH-inverted fluorescent in situ hybridization
- Probes used passed multiple specificity requirements involving both positive and negative controls (Table 6).
- Two jgISH probes were used: one that recognized gencDNAs via the exon16::exon 17 junction (Ex 16/17), which spans the Ab coding region of APP; and one that recognized IEJ formed between exons 3 and 16 (IEJ 3/16), representing one APP variant. All bound probes were enzymatically visualized, appearing as red dots (as indicated by the arrows) of varied diameter.
- jgISH probes produced similar results in RNase treated SAD neuronal nuclei ( FIGS. 19A-19B ).
- RNA signals were only detected using the anti-sense probes ( FIGS. 19C-19D ); therefore sense probes were exclusively used for genomic DNA detection.
- the jgISH sense probe signals were eliminated by specific restriction enzyme digestion of genomic DNA that eliminated the sequence recognition site ( FIGS. 19E-19J ).
- Non-classical gencDNA variants of APP were analyzed using multiple independent reactions on neuronal populations from brains ( FIG. 20A ), utilizing a DNA polymerase with 100 ⁇ higher fidelity compared to native Taq (Invitrogen, Platinum SuperFi DNA Polymerase). The resulting samples were pooled for library preparation to enable SMRT CCS of single DNA molecules. SMRT libraries yielded high-certainty consensus calling (20 CCS subreads with 99.9999% accuracy, median Phred score of 93). The number of unique sequences included 2,980 sequences. These included 21 different IEJs identified in neuronal nuclei of 2 SAD brains ( FIGS.
- FIGS. 20B-20E and 858 unique sequences including 11 IEJs in neuronal nuclei of 1 non-diseased brain. See FIG. 18C .
- GencDNAs of the canonical neuronal splice variant, APP 751 were also identified in both SAD and non-diseased datasets.
- SNVs, and insertions and deletions (INDELs) also occurred within APP gencDNAs of both SAD and non-diseased brain ( FIGS. 20E-20F ).
- GencDNA forms of mosaic neuronal recombination for potential relevance to SAD were assessed. Ten different SNVs in gencDNA variants from SAD but not non-diseased neurons were identified that were identical to pathogenic FAD APP mutations, including the Indiana mutation ( FIG. 20E and FIG. 21A ). Relationships of identified gencDNA variants to SAD were analyzed by comparing non-diseased and SAD neurons using jgISH.
- FIGS. 22A-22G The human transgene and non-classical variants of APP were analyzed. Predominant signals within neuronal nuclei, contrasting with low levels in non-neuronal nuclei from the same animals as well as wild-type controls were observed. See FIGS. 22A-22G .
- jgISH analyses of J20 neurons aged 177 vs. 829 days identified age-related increases in Ex 16/17 foci sizes in neurons ( FIGS. 23A-23C ). The size of foci reflected increased DNA copy number, as demonstrated by control experiments in which retroviral-mediated insertion of DNA target sequences with increasing 16/17 copies allowed semi-quantitation of jgISH foci sizes relative to target copy number ( FIGS. 23D-23F ).
- a ⁇ plaques were stained using an antibody recognizing A ⁇ peptide as seen in green in the first panel from the left.
- APP c-terminal (“c-terminal”) was stained using an antibody recognizing APP c-terminal as seen in red in arrows in the second panel from the left.
- DAPI (“DAPI”) was used to stain the nuclei as seen in blue in the fourth panel from the left.
- the data shows that non-classical variants of APP identified are present in A ⁇ plaques and are involved in A ⁇ plaque formation.
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Organic Chemistry (AREA)
- Genetics & Genomics (AREA)
- Engineering & Computer Science (AREA)
- Zoology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Molecular Biology (AREA)
- Wood Science & Technology (AREA)
- Biophysics (AREA)
- General Health & Medical Sciences (AREA)
- Biochemistry (AREA)
- Biomedical Technology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Biotechnology (AREA)
- General Engineering & Computer Science (AREA)
- Analytical Chemistry (AREA)
- Toxicology (AREA)
- Physics & Mathematics (AREA)
- Medicinal Chemistry (AREA)
- Microbiology (AREA)
- Gastroenterology & Hepatology (AREA)
- Neurology (AREA)
- Pathology (AREA)
- Immunology (AREA)
- Plant Pathology (AREA)
- Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
Abstract
Description
- This application claims benefit of U.S. Provisional Patent Application No. 62/500,270 filed on May 2, 2017, which is incorporated herein by reference in its entirety.
- The instant application contains a Sequence Listing which has been submitted electronically in ASCII format and is hereby incorporated by reference in its entirety. Said ASCII copy, created on May 1, 2018, is named 42256-722_601_SL.txt and is 28,069 bytes in size.
- Provided herein, in some embodiments, are methods for evaluating an individual for risk of developing a disease or disorder characterized by unwanted accumulation of amyloid beta protein comprising:
-
- (a) measuring an expression profile or an activity profile of one or more non-classical variant(s) of an amyloid beta precursor protein (APP) gene from a biological sample from the individual;
- (b) comparing the expression profile or the activity profile of the one or more non-classical variant(s) to a reference expression profile or activity profile of the one or more non-classical variant(s) derived from a cohort of control individuals; and
- (c) identifying the individual as having or not having a risk factor for developing the disease or disorder characterized by unwanted accumulation of amyloid beta protein based on a comparison of the expression profile or the activity profile measured in step (a) to the reference expression profile or activity profile of the cohort of control individuals.
In some embodiments, the expression profile is expression level of the one or more non-classical variant(s). In some embodiments, the activity profile is activity level of the one or more non-classical variant(s). In some embodiments, the expression profile is expression of a set of different non-classical variants. In some embodiments, the activity profile is activity of a set of different non-classical variants. In some embodiments, methods further comprise measuring the expression profile by a method comprising long-read sequencing of the biological sample from the individual. In some embodiments, the long-read sequencing is RNA sequencing (RNA-seq). In some embodiments, the long-read sequencing is DNA sequencing. In some embodiments, the one or more non-classical variant(s) comprises a portion or all ofexon exon 8. In some embodiments, one or more exon(s) in the one or more non-classical variant(s) is rearranged as compared to a control. In some embodiments, the one or more non-classical variant(s) comprises a single nucleotide variation (SNV) in APP. In some embodiments, the SNV in APP translates to amino acid positions in APP selected from a group consisting of A673V, A713T, T714I, V715M, V715A, I716M, V717I, V717F, T719P, and L723P. In some embodiments, generation of the one or more non-classical variant(s) involves transcription. In some embodiments, the one or more non-classical variant(s) comprises a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16. In some embodiments, the disease or disorder is Alzheimer's disease. In some embodiments, Alzheimer's disease is familial Alzheimer's disease (FAD) or sporadic Alzheimer's disease. In some embodiments, the biological sample is collected from blood or cerebrospinal fluid. In some embodiments, the biological sample comprises RNA, DNA, or protein. In some embodiments, the DNA is genomic DNA, extrachromosomal DNA, or circular DNA. In some embodiments, the DNA is genomic DNA. In some embodiments, methods further comprise isolating and purifying RNA or DNA from the biological sample prior to the long-read sequencing. In some embodiments, methods further comprise reverse transcribing RNA to cDNA prior to the long-read sequencing. In some embodiments, an average read length for the long-read sequencing is at least 5000 bases. In some embodiments, measuring the expression profile comprises quantifying the expression level of the one or more non-classical variant(s) by a method comprising counting a number of reads that map to a sequence of the one or more non-classical variant(s). In some embodiments, measuring the expression profile comprises a method comprising binding of one or more probe(s) to the one or more non-classical variant(s). In some embodiments, the expression profile comprises quantifying the expression level by a method comprising quantitative polymerase chain reaction (qPCR). In some embodiments, the one or more probe(s) are selected from polynucleotides or polypeptides. In some embodiments, the one or more probe(s) hybridize to RNA or DNA within the biological sample. In some embodiments, the one or more probe(s) hybridize to a range of about 35 to about 50 nucleotides in the RNA or DNA. In some embodiments, methods further comprise capturing the one or more non-classical variant(s) from the biological sample on a solid support prior to contacting the one or more non-classical variant(s) with the one or more probe(s). In some embodiments, the one or more non-classical variant(s) is detected by in situ hybridization or immunological hybridization. In some embodiments, the in situ hybridization is chromogenic in situ hybridization or fluorescence in situ hybridization. In some embodiments, measuring binding of the one or more probe(s) to the one or more non-classical variant(s) further comprises a pull-down assay. In some embodiments, the one or more probe(s) for the pull-down assay are designed to hybridize to a portion or all ofexon
- Provided herein, in some embodiments, are methods of treating a disease or disorder characterized by unwanted accumulation of amyloid beta protein in an individual in need thereof, comprising: administering to the individual an agent that inhibits activity of one or more non-classical variant(s) of an APP gene. In some embodiments, the one or more non-classical variant(s) comprises a portion or all of
exon exon 8. In some embodiments, one or more exon(s) in the one or more non-classical variant(s) is rearranged as compared to a control. In some embodiments, the one or more non-classical variant(s) comprises a single nucleotide variation (SNV) in APP. In some embodiments, the SNV in APP translates to amino acid positions in APP selected from a group consisting of A673V, A713T, T714I, V715M, V715A, I716M, V717I, V717F, T719P, and L723P. In some embodiments, generation of the one or more non-classical variant(s) involves transcription. In some embodiments, the one or more non-classical variant(s) comprises a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16. In some embodiments, the disease or disorder is Alzheimer's disease. In some embodiments, Alzheimer's disease is familial Alzheimer's disease (FAD) or sporadic Alzheimer's disease. In some embodiments, the agent that inhibits the activity of APP is an antibody, an antigen binding fragment, a RNA interfering agent (RNAi), a small interfering RNA (siRNA), a short hairpin RNA (shRNA), a microRNA (miRNA), an antisense oligonucleotide, a peptide, a peptidomimetic, a small molecule, or an aptamer. In some embodiments, the antibody binds to a protein encoded by the one or more non-classical variant(s). In some embodiments, the antisense oligonucleotide targets RNA or DNA of APP. In some embodiments, the RNA is mRNA. In some embodiments, the DNA is genomic DNA. In some embodiments, the protein is encoded by the one or more non-classical variant(s) comprising a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16. In some embodiments, the agent does not inhibit activity of a wild-type amyloid beta precursor protein gene or protein thereof. In some embodiments, the unwanted accumulation of amyloid beta protein is plaque depositions comprising a portion of or all amyloid beta protein, or accumulation of soluble amyloid beta protein. In some embodiments, the unwanted accumulation of amyloid beta protein results from increased APP associated with the one or more non-classical variant(s) of APP gene. - Provided herein, in some embodiments, are methods of diagnosing and treating a disease or disorder characterized by unwanted accumulation of amyloid beta protein in an individual in need thereof, comprising:
-
- (a) identifying the individual as having the disease or disorder characterized by unwanted accumulation of amyloid beta protein if binding of one or more probe(s) to one or more non-classical variant(s) of an APP gene is measured; and
- (b) administering to the individual having an expression profile or an activity profile of the one or more non-classical variant(s) an agent that inhibits activity of the one or more non-classical variant(s).
In some embodiments, methods further comprise contacting a biological sample from the individual with the one or more probe(s) that hybridize to the one or more non-classical variant(s). In some embodiments, methods further comprise measuring binding of the one or more probe(s) to the one or more non-classical variant(s). In some embodiments, the expression profile is expression level of the one or more non-classical variant(s). In some embodiments, the activity profile is activity level of the one or more non-classical variant(s). In some embodiments, methods further comprise quantifying the expression level of the one or more non-classical variant(s) by a method comprising quantitative polymerase chain reaction (qPCR). In some embodiments, the expression profile is expression of a set of different non-classical variants. In some embodiments, the activity profile is activity of a set of different non-classical variants. In some embodiments, the one or more non-classical variant(s) comprises a portion or all ofexon exon 8. In some embodiments, one or more exon(s) in the one or more non-classical variant(s) is rearranged as compared to a control. In some embodiments, the one or more non-classical variant(s) comprises a single nucleotide variation (SNV) in APP. In some embodiments, the SNV in APP translates to amino acid positions in APP selected from a group consisting of A673V, A713T, T714I, V715M, V715A, I716M, V717I, V717F, T719P, and L723P. In some embodiments, generation of the one or more non-classical variant(s) involves transcription. In some embodiments, the one or more non-classical variant(s) comprises a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16. In some embodiments, the disease or disorder is Alzheimer's disease. In some embodiments, Alzheimer's disease is familial Alzheimer's disease (FAD) or sporadic Alzheimer's disease. In some embodiments, the biological sample is collected from blood or cerebrospinal fluid. In some embodiments, the biological sample comprises RNA, DNA, or protein. In some embodiments, the DNA is genomic DNA, extrachromosomal DNA, or circular DNA. In some embodiments, the DNA is genomic DNA. In some embodiments, the one or more probe(s) are selected from polynucleotides or polypeptides. In some embodiments, the one or more probe(s) hybridize to RNA or DNA within the biological sample. In some embodiments, the one or more probe(s) hybridize to a range of about 35 to about 50 nucleotides of the RNA or DNA. In some embodiments, methods further comprise capturing the one or more non-classical variant(s) from the biological sample prior to contacting the one or more non-classical variant(s) with the one or more probe(s). In some embodiments, the one or more non-classical variant(s) is detected by in situ hybridization or immunological hybridization. In some embodiments, the in situ hybridization is chromogenic in situ hybridization or fluorescence in situ hybridization. In some embodiments, methods further comprise measuring binding of the one or more probe(s) to the one or more non-classical variant(s) further comprises a pull-down assay. In some embodiments, the one or more probe(s) for the pull-down assay are designed to hybridize to a portion or all ofexon
- Provided herein, in some embodiments, are methods of diagnosing and treating a disease or disorder characterized by unwanted accumulation of amyloid beta protein in an individual in need thereof, comprising:
-
- (a) identifying the individual as having the disease or disorder characterized by unwanted accumulation of amyloid beta protein if an expression profile of one or more non-classical variant(s) of an APP gene is measured by a method comprising long-read sequencing of a biological sample from the individual; and
- (b) administering to the individual having the expression profile of the one or more non-classical variant(s) an agent that inhibits activity of the one or more non-classical variant(s).
In some embodiments, the expression profile is expression level of the one or more non-classical variant(s). In some embodiments, the long-read sequencing is RNA sequencing (RNA-seq). In some embodiments, the long-read sequencing is DNA sequencing. In some embodiments, methods further comprise quantifying the expression level of the one or more non-classical variant(s) by a method comprising counting a number of reads that map to a sequence of the one or more non-classical variant(s). In some embodiments, the expression profile is expression of a set of different non-classical variants. In some embodiments, the one or more non-classical variant(s) comprises a portion or all ofexon exon 8. In some embodiments, one or more exon(s) in the one or more non-classical variant(s) is rearranged as compared to a control. In some embodiments, the one or more non-classical variant(s) comprises a single nucleotide variation (SNV) in APP. In some embodiments, the SNV in APP translates to amino acid positions in APP selected from a group consisting of A673V, A713T, T714I, V715M, V715A, I716M, V717I, V717F, T719P, and L723P. In some embodiments, generation of the one or more non-classical variant(s) involves transcription. In some embodiments, the one or more non-classical variant(s) comprises a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16. In some embodiments, the disease or disorder is Alzheimer's disease. In some embodiments, Alzheimer's disease is familial Alzheimer's disease (FAD) or sporadic Alzheimer's disease. In some embodiments, the biological sample is collected from blood or cerebrospinal fluid. In some embodiments, the biological sample comprises RNA or DNA. In some embodiments, the DNA is genomic DNA, extrachromosomal DNA, or circular DNA. In some embodiments, the DNA is genomic DNA. In some embodiments, methods further comprise isolating and purifying RNA or DNA from the biological sample prior to the long-read sequencing. In some embodiments, methods further comprise reverse transcribing RNA to cDNA prior to the long-read sequencing. In some embodiments, an average read length for the long-read sequencing is at least 5000 bases. In some embodiments, the agent that inhibits the activity of APP is an antibody, an antigen binding fragment, a RNA interfering agent (RNAi), a small interfering RNA (siRNA), a short hairpin RNA (shRNA), a microRNA (miRNA), an antisense oligonucleotide, a peptide, a peptidomimetic, a small molecule, or an aptamer. In some embodiments, the unwanted accumulation of amyloid beta protein is plaque depositions comprising a portion of or all amyloid beta protein, or accumulation soluble amyloid beta protein. In some embodiments, the antisense oligonucleotide targets RNA or DNA of APP. In some embodiments, the RNA is mRNA. In some embodiments, the DNA is genomic DNA.
- Provided herein, in some embodiments, are methods of detecting one or more non-classical variant(s) of an APP gene in an individual in need thereof, comprising: detecting an expression profile of the one or more non-classical variant(s) of the APP gene in a biological sample from the individual by a method comprising long-read sequencing of the biological sample. In some embodiments, the expression profile is expression level of the one or more non-classical variant(s). In some embodiments, the expression profile is expression of a set of different non-classical variants. In some embodiments, the one or more non-classical variant(s) comprises a portion or all of
exon exon 8. In some embodiments, one or more exon(s) in the one or more non-classical variant(s) is rearranged as compared to a control. In some embodiments, the one or more non-classical variant(s) comprises a single nucleotide variation (SNV) in APP. In some embodiments, the SNV in APP translates to amino acid positions in APP selected from a group consisting of A673V, A713T, T714I, V715M, V715A, I716M, V717I, V717F, T719P, and L723P. In some embodiments, generation of the one or more non-classical variant(s) involves transcription. In some embodiments, the one or more non-classical variant(s) comprises a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16. In some embodiments, the expression profile of the one or more non-classical variant(s) is associated with a disease or disorder characterized by unwanted accumulation of amyloid beta protein. In some embodiments, the unwanted accumulation of amyloid beta protein is plaque depositions comprising a portion of or all amyloid beta protein, or accumulation of soluble amyloid beta protein. In some embodiments, the expression profile of the one or more non-classical variant(s) is associated with unwanted accumulation of amyloid beta protein, and wherein the individual does not have a disease or disorder. In some embodiments, the disease or disorder is Alzheimer's disease. In some embodiments, the biological sample is collected from blood or cerebrospinal fluid. In some embodiments, the biological sample comprises RNA or DNA. In some embodiments, the DNA is genomic DNA, extrachromosomal DNA, or circular DNA. In some embodiments, the DNA is genomic DNA. In some embodiments, methods further comprise isolating and purifying RNA or DNA from the biological sample prior to the long-read sequencing. In some embodiments, methods further comprise reverse transcribing RNA to cDNA prior to the long-read sequencing. In some embodiments, an average read length for the long-read sequencing is at least 5000 bases. In some embodiments, the long-read sequencing is RNA sequencing (RNA-seq). In some embodiments, the long-read sequencing is DNA sequencing. In some embodiments, detecting the expression profile comprises quantifying the expression level of the one or more non-classical variant(s) by a method comprising counting a number of reads that map to a sequence of the one or more non-classical variant(s). In some embodiments, the individual is suspected of having or being predisposed to Alzheimer's disease. - Provided herein, in some embodiments, are methods of detecting one or more non-classical variant(s) of an APP gene in an individual in need thereof, comprising: detecting an expression profile or an activity profile of the one or more non-classical variant of the APP gene in a biological sample from the individual by a method comprising binding of one or more probe(s) to the one or more non-classical variant(s). In some embodiments, the expression profile is expression level of the one or more non-classical variant(s). In some embodiments, the activity profile is activity level of the one or more non-classical variant(s). In some embodiments, the expression profile is expression of a set of different non-classical variants. In some embodiments, the activity profile is activity of a set of different non-classical variants. In some embodiments, detecting the expression profile comprises quantifying expression level by a method comprising quantitative polymerase chain reaction (qPCR). In some embodiments, the one or more non-classical variant(s) comprises a portion or all of
exon exon 8. In some embodiments, one or more exon(s) in the one or more non-classical variant(s) is rearranged as compared to a control. In some embodiments, the one or more non-classical variant(s) comprises a single nucleotide variation (SNV) in APP. In some embodiments, the SNV in APP translates to amino acid positions in APP selected from a group consisting of A673V, A713T, T714I, V715M, V715A, I716M, V717I, V717F, T719P, and L723P. In some embodiments, generation of the one or more non-classical variant(s) involves transcription. In some embodiments, the one or more non-classical variant(s) comprises a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16. In some embodiments, the expression profile or the activity level of the one or more non-classical variant(s) is associated with a disease or disorder characterized by unwanted accumulation of amyloid beta protein. In some embodiments, the unwanted accumulation of amyloid beta protein is plaque depositions comprising a portion of or all amyloid beta protein, or accumulation of soluble amyloid beta protein. In some embodiments, the expression profile or the activity profile of the one or more non-classical variant(s) is associated with unwanted accumulation of amyloid beta protein, and wherein the individual does not have a disease or disorder. In some embodiments, the disease or disorder is Alzheimer's disease. In some embodiments, the biological sample is collected from blood or cerebrospinal fluid. In some embodiments, the biological sample comprises RNA, DNA, or protein. In some embodiments, the DNA is genomic DNA, extrachromosomal DNA, or circular DNA. In some embodiments, the DNA is genomic DNA. In some embodiments, the one or more probe(s) are selected from polynucleotides or polypeptides. In some embodiments, the one or more probe(s) hybridize to RNA or DNA within the biological sample. In some embodiments, the one or more probe(s) hybridize to a range of about 35 to about 50 nucleotides of the RNA or DNA. In some embodiments, methods further comprise capturing the one or more non-classical variant(s) from the biological sample on a solid support prior to contacting the one or more non-classical variant with the one or more probe(s). In some embodiments, the one or more non-classical variant(s) is detected by in situ hybridization or immunological hybridization. In some embodiments, the in situ hybridization is chromogenic in situ hybridization or fluorescence in situ hybridization. In some embodiments, detecting binding of the one or more probe(s) to the one or more non-classical variant(s) further comprises a pull-down assay. In some embodiments, the one or more probe(s) for the pull-down assay are designed to hybridize to a portion or all ofexon - Provided herein, in some embodiments, are methods of diagnosing a disease or disorder in an individual characterized by unwanted accumulation of amyloid beta protein, comprising: identifying the individual as having the disease or disorder characterized by unwanted accumulation of amyloid beta protein by comparing an expression profile of one or more non-classical variant(s) of an APP gene to a reference expression profile of the one or more non-classical variant(s) derived from a cohort of control individuals, wherein the expression profile of the one or more non-classical variant(s) is measured by a method comprising long-read sequencing of a biological sample from the individual; and wherein the expression profile of the one or more non-classical variant(s) is associated with the disease or disorder. In some embodiments, the expression profile is expression level of the one or more non-classical variant(s). In some embodiments, the expression profile is expression of a set of different non-classical variants. In some embodiments, the long-read sequencing is RNA sequencing (RNA-seq). In some embodiments, the long-read sequencing is DNA sequencing. In some embodiments, methods further comprise quantifying the expression level of the one or more non-classical variant(s) by a method comprising counting a number of reads that map to a sequence of the one or more non-classical variant(s). In some embodiments, the one or more non-classical variant(s) comprises a portion or all of
exon exon 8. In some embodiments, one or more exon(s) in the one or more non-classical variant(s) is rearranged as compared to a control. In some embodiments, the one or more non-classical variant(s) comprises a single nucleotide variation (SNV) in APP. In some embodiments, the SNV in APP translates to amino acid positions in APP selected from a group consisting of A673V, A713T, T714I, V715M, V715A, I716M, V717I, V717F, T719P, and L723P. In some embodiments, generation of the one or more non-classical variant(s) involves transcription. In some embodiments, the one or more non-classical variant(s) comprises a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16. In some embodiments, the disease or disorder is Alzheimer's disease. In some embodiments, Alzheimer's disease is familial Alzheimer's disease (FAD) or sporadic Alzheimer's disease. In some embodiments, the individual is suspected of having or being predisposed to Alzheimer's disease. In some embodiments, the biological sample is collected from blood or cerebrospinal fluid. In some embodiments, methods further comprise isolating and purifying RNA or DNA from the biological sample prior to the long-read sequencing. In some embodiments, methods further comprise reverse transcribing RNA to cDNA prior to the long-read sequencing. In some embodiments, an average read length for the long-read sequencing is at least 5000 bases. In some embodiments, the unwanted accumulation of amyloid beta protein is plaque depositions comprising a portion of or all amyloid beta protein, or accumulation of soluble amyloid beta protein. - Provided herein, in some embodiments, are methods of diagnosing a disease or disorder in an individual characterized by unwanted accumulation of amyloid beta protein, comprising: identifying the individual as having the disease or disorder characterized by unwanted accumulation of amyloid beta protein by comparing an expression profile or an activity profile of one or more non-classical variant(s) of an APP gene to a reference expression profile of the one or more non-classical variant(s) derived from a cohort of control individuals, wherein the expression profile or the activity profile of the one or more non-classical variant(s) is measured by a method comprising binding of one or more probe(s) to a biological sample from the individual; and wherein the expression profile or the activity profile of the one or more non-classical variant(s) is associated with the disease or disorder. In some embodiments, the expression profile is expression level of the one or more non-classical variant(s). In some embodiments, the activity profile is activity level of the one or more non-classical variant(s). In some embodiments, the expression profile is expression of a set of different non-classical variants. In some embodiments, the activity profile is activity of a set of different non-classical variants. In some embodiments, methods further comprise quantifying the expression level by a method comprising quantitative polymerase chain reaction (qPCR). In some embodiments, methods further comprise capturing the one or more non-classical variant(s) from the biological sample on a solid support prior to contacting the one or more non-classical variant(s) with the one or more probe(s). In some embodiments, the one or more non-classical variant(s) is detected by in situ hybridization or immunological hybridization. In some embodiments, the in situ hybridization is chromogenic in situ hybridization or fluorescence in situ hybridization. In some embodiments, binding of the one or more probe(s) to the one or more non-classical variant(s) further comprises a pull-down assay. In some embodiments, the one or more probe(s) for the pull-down assay are designed to hybridize to a portion or all of
exon - Provided herein, in some embodiments, are kits for detecting one or more non-classical variant(s) of an APP gene in a biological sample of an individual, the kit comprising:
-
- (a) one or more probe(s) that hybridize to the one or more non-classical variant(s) of the APP gene or protein thereof; and
- (b) a detecting reagent for examining binding of the one or more probe(s) with the one or more non-classical variant(s) of the APP gene or protein thereof.
In some embodiments, the one or more non-classical variant(s) comprises a portion or all ofexon exon 8. In some embodiments, one or more exon(s) in the one or more non-classical variant(s) is rearranged as compared to a control. In some embodiments, the one or more non-classical variant(s) comprises a single nucleotide variation (SNV) in APP. In some embodiments, the SNV in APP translates to amino acid positions in APP selected from a group consisting of A673V, A713T, T714I, V715M, V715A, I716M, V717I, V717F, T719P, and L723P. In some embodiments, generation of the one or more non-classical variant(s) involves transcription. In some embodiments, the one or more non-classical variant(s) comprises a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16. In some embodiments, the one or more probe(s) are selected from polynucleotides or polypeptides. In some embodiments, the one or more probe(s) hybridize to RNA or DNA within the biological sample. In some embodiments, the one or more probe(s) bind to one or more protein(s) encoded by the one or more non-classical variant(s). In some embodiments, the one or more probe(s) comprise an antibody or fragment thereof. In some embodiments, a probe in the one or more probe(s) is labeled using a fluorochrome or a radioactive isotope. In some embodiments, a probe in the one or more probe(s) is labeled using an affinity tag. In some embodiments, the affinity tag is biotin, desthiobiotin, histidine, polyhistidine, myc, hemagglutinin (HA), FLAG, glutathione S transferase (GST), or derivatives thereof. In some embodiments, the affinity tag is recognized by avidin, streptavidin, nickel, or glutathione. In some embodiments, the detecting reagent binds to the one or more probe(s). In some embodiments, the detecting reagent comprises a fluorescent or a radioactive label. In some embodiments, the individual is suspected of having or being predisposed to Alzheimer's disease.
- Provided herein, in some embodiments, are in vitro methods of screening for a therapeutic agent for treating a disease or disorder characterized by unwanted accumulation of amyloid beta protein, comprising:
-
- (a) contacting a cell that expresses a non-classical variant of an APP gene with a test agent;
- (b) detecting inhibition of expression of the non-classical variant of the APP gene compared to a control; and
- (c) identifying the test agent as a therapeutic agent if the test agent inhibits expression of the non-classical variant of the APP gene compared to the control.
In some embodiments, expression of the non-classical variant of the APP gene is measured by qPCR or gel electrophoresis. In some embodiments, detecting inhibition of expression comprises measuring protein expression of a protein encoded by the non-classical variant of the APP gene. In some embodiments, the protein expression is measured by Western blot, enzyme-linked immunosorbent assays (ELISA), or chromatography.
- Provided herein, in some embodiments, are in vitro methods of screening for a therapeutic agent for treating a disease or disorder characterized by unwanted accumulation of amyloid beta protein, comprising:
-
- (a) contacting a cell that expresses a non-classical variant of an APP gene with a test agent;
- (b) detecting inhibition of activity of the non-classical variant of the APP gene as compared to a control; and
- (c) identifying the test agent as the therapeutic agent if the test agent inhibits the activity of the non-classical variant of the APP gene as compared to the control.
In some embodiments, the activity of the non-classical variant comprises accumulation of amyloid beta protein. In some embodiments, the accumulation of amyloid beta protein is measured by a method comprising Western blot, enzyme-linked immunosorbent assays (ELISA), or chromatography. In some embodiments, the activity of the non-classical variant is measured by a method comprising a fluorescence assay, a luciferase assay, or an enzymatic assay.
- Provided herein, in some embodiments, are in vitro methods of screening for a therapeutic agent for treating a disease or disorder characterized by unwanted accumulation of amyloid beta protein, comprising:
-
- (a) contacting a cell that expresses a non-classical variant of an APP gene with a test agent;
- (b) detecting binding of the test agent to the non-classical variant of the APP gene; and
- (c) identifying the test agent as the therapeutic agent if the test agents binds to the non-classical variant of the APP gene.
In some embodiments, methods further comprise detecting binding by a method comprising a radioactive binding assay, a fluorescence binding assay, an enzyme-linked immunosorbent assay (ELISA), a kinetic exclusion assay, or a crystallography assay. In some embodiments, the non-classical variant comprises a portion or all ofexon exon 8. In some embodiments, one or more exon(s) in the non-classical variant is rearranged as compared to a control. In some embodiments, the one or more non-classical variant(s) comprises a single nucleotide variation (SNV) in APP. In some embodiments, the SNV in APP translates to amino acid positions in APP selected from a group consisting of A673V, A713T, T714I, V715M, V715A, I716M, V717I, V717F, T719P, and L723P. In some embodiments, the non-classical variant comprises a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16. In some embodiments, the therapeutic agent is an antibody, an antigen binding fragment, a RNA interfering agent (RNAi), a small interfering RNA (siRNA), a short hairpin RNA (shRNA), a microRNA (miRNA), an antisense oligonucleotide, a peptide, a peptidomimetic, a small molecule, or an aptamer. In some embodiments, the antisense oligonucleotide targets RNA or DNA of APP. In some embodiments, the RNA is mRNA. In some embodiments, the DNA is genomic DNA.
- Provided herein, in some embodiments, are methods of identifying one or more non-classical variant(s) of an APP gene, comprising:
-
- (a) isolating RNA from a cell from an individual with Alzheimer's disease;
- (b) reverse transcribing the RNA into cDNA;
- (c) hybridizing the cDNA with a probe to detect APP; and
- (d) sequencing the cDNA that was detected by step (c).
In some embodiments, the probe comprises a sequence as set forth in SEQ ID NO: 21. In some embodiments, the cell is a neuron. In some embodiments, methods further comprise comparing cDNA isolated from RNA of an individual without Alzheimer's disease.
- Provided herein, in some embodiments, are therapeutic agents identified by any of the methods described herein.
- Provided herein, in some embodiments, are methods of treating a disease or disorder characterized by unwanted accumulation of amyloid beta protein in an individual in need thereof, comprising: administering to the individual an agent that inhibits generation of one or more non-classical variant(s) of an APP gene. In some embodiments, the generation of the one or more non-classical variant(s) involves transcription. In some embodiments, the agent edits RNA. In some embodiments, the agent edits DNA. In some embodiments, the one or more non-classical variant(s) comprises a portion or all of
exon exon 8. In some embodiments, one or more exon(s) in the one or more non-classical variant(s) is rearranged as compared to a control. In some embodiments, the one or more non-classical variant(s) comprises a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16. In some embodiments, the disease or disorder is Alzheimer's disease. In some embodiments, Alzheimer's disease is familial Alzheimer's disease (FAD) or sporadic Alzheimer's disease. In some embodiments, the unwanted accumulation of amyloid beta protein is plaque depositions comprising a portion of or all amyloid beta protein, or accumulation of soluble amyloid beta protein. In some embodiments, the unwanted accumulation of amyloid beta protein results from increased APP associated with the one or more non-classical variant(s) of APP gene. - Provided herein, in some embodiments, are methods of treating a disease or disorder characterized by unwanted accumulation of amyloid beta protein in an individual in need thereof, comprising: administering to the individual a therapeutically-effective amount of a composition comprising: (a) a nuclease; and (b) a guide nucleic acid comprising a region that is complementary to a target nucleic acid, wherein the target nucleic acid is a non-classical variant of APP. In some embodiments, the nuclease is a CRISPR-associated protein (Cas). In some embodiments, the Cas protein is Cas9. In some embodiments, the Cas protein is Cas13. In some embodiments, the target nucleic acid is RNA of the non-classical variant of APP. In some embodiments, the target nucleic acid is DNA of the non-classical variant of APP. In some embodiments, the non-classical variant comprises a portion or all of
exon exon 8. In some embodiments, one or more exon(s) in the non-classical variant is rearranged as compared to a control. In some embodiments, the non-classical variant comprises a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16. In some embodiments, the disease or disorder is Alzheimer's disease. In some embodiments, Alzheimer's disease is familial Alzheimer's disease (FAD) or sporadic Alzheimer's disease. In some embodiments, the unwanted accumulation of amyloid beta protein is plaque depositions comprising a portion of or all amyloid beta protein, or accumulation of soluble amyloid beta protein. In some embodiments, unwanted accumulation of amyloid beta protein results from increased APP associated with the non-classical variant of APP gene. - Provided herein, in some embodiments, are methods method of detecting one or more non-classical variant(s) of an APP gene in an individual in need thereof, comprising: detecting an expression profile of the one or more non-classical variant(s) of the APP gene in a biological sample from the individual by (a) long-read sequencing of RNA or DNA encoding for the one or more non-classical variant(s), or (b) binding of one or more probe(s) to the one or more non-classical variant(s). In some embodiments, the expression profile is expression level of the one or more non-classical variant(s) or expression of a set of different non-classical variants. In some embodiments, the one or more non-classical variant(s) comprises a portion or all of
exon exon 8. In some embodiments, one or more exon(s) in the one or more non-classical variant(s) is rearranged as compared to a control. In some embodiments, the one or more non-classical variant(s) comprises a single nucleotide variation (SNV) in APP. In some embodiments, the SNV in APP translates to amino acid positions in APP selected from a group consisting of A673V, A713T, T714I, V715M, V715A, I716M, V717I, V717F, T719P, and L723P. In some embodiments, the one or more non-classical variant(s) comprises a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16. In some embodiments, the expression profile of the one or more non-classical variant(s) is associated with a disease or disorder characterized by unwanted accumulation of amyloid beta protein. In some embodiments, the unwanted accumulation of amyloid beta protein is plaque depositions comprising a portion of or all amyloid beta protein, or accumulation of soluble amyloid beta protein. In some embodiments, the expression profile of the one or more non-classical variant(s) is associated with unwanted accumulation of amyloid beta protein, and wherein the individual does not have a disease or disorder. In some embodiments, the biological sample is blood or cerebrospinal fluid. In some embodiments, the DNA is genomic DNA, extrachromosomal DNA, or circular DNA. In some embodiments, detecting the expression profile comprises quantifying the expression level of the one or more non-classical variant(s) by a method comprising counting a number of reads that map to a sequence of the one or more non-classical variant(s). In some embodiments, the individual is suspected of having or being predisposed to Alzheimer's disease. In some embodiments, the one or more probe(s) hybridize to RNA or DNA encoding for the one or more non-classical variant(s) within the biological sample. In some embodiments, the one or more probe(s) hybridize to an intraexonic junction betweenexon 1 andexon 11,exon 1 andexon 14,exon 2 andexon 17,exon 2 andexon 14,exon 2 andexon 18,exon 2 andexon 16,exon 3 andexon 16,exon 3 andexon 14,exon 3 andexon 17,exon 6 andexon 17,exon 6 andexon 18,exon 3 andexon 9,exon 2 andexon 9,exon 16 andexon 18,exon 6 andexon 12,exon 5 andexon 16, orexon 16 andexon 17. In some embodiments, the one or more non-classical variant(s) is detected by in situ hybridization or immunological hybridization. In some embodiments, detecting binding of the one or more probe(s) to the one or more non-classical variant(s) further comprises a pull-down assay. In some embodiments, the one or more probe(s) for the pull-down assay are designed to hybridize to a portion or all ofexon - Provided herein, in some embodiments, are methods for evaluating an individual for risk of developing a disease or disorder characterized by unwanted accumulation of amyloid beta protein comprising:
-
- (a) measuring an expression profile or an activity profile of one or more non-classical variant(s) of an amyloid beta precursor protein (APP) gene from a biological sample from the individual;
- (b) comparing the expression profile or the activity profile of the one or more non-classical variant(s) to a reference expression profile or activity profile of the one or more non-classical variant(s) derived from a cohort of control individuals; and
- (c) identifying the individual as having or not having a risk factor for developing the disease or disorder characterized by unwanted accumulation of amyloid beta protein based on a comparison of the expression profile or the activity profile measured in step (a) to the reference expression profile or activity profile of the cohort of control individuals.
Provided herein, in some embodiments, are methods of diagnosing a disease or disorder characterized by unwanted accumulation of amyloid beta protein in an individual in need thereof, comprising: - (a) measuring an expression profile or an activity profile of one or more non-classical variant(s) of an amyloid beta precursor protein (APP) gene from a biological sample from the individual by (i) long-read sequencing of RNA or DNA encoding for the one or more non-classical variant(s), or (ii) binding of one or more probe(s) to the one or more non-classical variant(s); and
- (b) identifying the individual as having the disease or disorder characterized by unwanted accumulation of amyloid beta protein by comparing the expression profile or the activity profile of one or more non-classical variant(s) of an APP gene to a reference expression profile or an activity profile of the one or more non-classical variant(s) derived from a cohort of control individuals,
- wherein the expression profile or the activity profile of the one or more non-classical variant(s) is associated with the disease or disorder.
- In some embodiments, the expression profile is expression level of the one or more non-classical variant(s), and wherein the activity profile is activity level of the one or more non-classical variant(s). In some embodiments, the expression profile is expression of a set of different non-classical variants, and wherein the activity profile is activity of a set of different non-classical variants. In some embodiments, methods further comprise measuring the expression profile by a method comprising long-read sequencing of RNA or DNA from the biological sample from the individual, wherein the RNA or DNA encodes the one or more non-classical variant(s). In some embodiments, measuring the expression profile comprises quantifying the expression level of the one or more non-classical variant(s) by a method comprising counting a number of reads that map to a sequence of the one or more non-classical variant(s). In some embodiments, the one or more non-classical variant(s) comprises a portion or all of
exon exon 8. In some embodiments, one or more exon(s) in the one or more non-classical variant(s) is rearranged as compared to a control. In some embodiments, the one or more non-classical variant(s) comprises a single nucleotide variation (SNV) in APP. In some embodiments, the SNV in APP translates to amino acid positions in APP selected from a group consisting of A673V, A713T, T714I, V715M, V715A, I716M, V717I, V717F, T719P, and L723P. In some embodiments, the one or more non-classical variant(s) comprises a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16. In some embodiments, the disease or disorder is Alzheimer's disease. In some embodiments, the Alzheimer's disease is familial Alzheimer's disease (FAD) or sporadic Alzheimer's disease. In some embodiments, the biological sample is blood or cerebrospinal fluid. In some embodiments, the biological sample comprises RNA, DNA, or protein encoding for the one or more non-classical variant(s). In some embodiments, the DNA is genomic DNA, extrachromosomal DNA, or circular DNA. In some embodiments, measuring the expression profile comprises a method comprising binding of one or more probe(s) to the one or more non-classical variant(s). In some embodiments, the one or more probe(s) hybridize to an intraexonic junction betweenexon 1 andexon 11,exon 1 andexon 14,exon 2 andexon 17,exon 2 andexon 14,exon 2 andexon 18,exon 2 andexon 16,exon 3 andexon 16,exon 3 andexon 14,exon 3 andexon 17,exon 6 andexon 17,exon 6 andexon 18,exon 3 andexon 9,exon 2 andexon 9,exon 16 andexon 18,exon 6 andexon 12,exon 5 andexon 16, orexon 16 andexon 17. In some embodiments, measuring the expression profile comprises quantifying the expression level by a method comprising quantitative polymerase chain reaction (qPCR). In some embodiments, the one or more probe(s) hybridize to RNA or DNA encoding for the one or more non-classical variant(s) within the biological sample. In some embodiments, the one or more non-classical variant(s) is detected by in situ hybridization or immunological hybridization. In some embodiments, measuring binding of the one or more probe(s) to the one or more non-classical variant(s) further comprises a pull-down assay. In some embodiments, the one or more probe(s) for the pull-down assay are designed to hybridize to a portion or all ofexon - Provided herein, in some embodiments, are methods of treating a disease or disorder characterized by unwanted accumulation of amyloid beta protein in an individual in need thereof, comprising:
-
- (a) performing (i) long-read sequencing of RNA or DNA encoding one or more non-classical variant(s) of an APP gene in a biological sample or (ii) a binding assay using one or more probe(s) to the one or more non-classical variant(s);
- (b) selecting the individual as having the disease or disorder characterized by unwanted accumulation of amyloid beta protein who has an elevated expression profile or an elevated activity profile of the one or more non-classical variant(s) of an APP gene compared to a reference expression profile or reference activity profile of the one or more non-classical variant(s) derived from a cohort of control individuals; and
- (c) administering to the individual having the elevated expression profile or the elevated activity profile of the one or more non-classical variant(s) an agent that inhibits activity of the one or more non-classical variant(s).
Provided herein, in some embodiments, are methods of treating Alzheimer's disease in an individual, comprising: - (a) obtaining an expression profile or an activity profile of the one or more non-classical variant(s) of an APP gene from a biological sample from the individual, wherein the expression profile or the activity profile is determined by (i) long-read sequencing of RNA or DNA encoding for the one or more non-classical variant(s), or (ii) binding of one or more probe(s) to the one or more non-classical variant(s);
- (b) determining the individual has or is predisposed to Alzheimer's disease based on the expression profile or the activity profile from (a), wherein a likelihood of having or being predisposed to Alzheimer's disease is increased when the expression profile or the activity profile is elevated compared to a reference expression profile or reference activity profile of the one or more non-classical variant(s) derived from a cohort of control individuals; and
- (c) treating the individual for Alzheimer's disease following (b).
Provided herein, in some embodiments, are methods for identifying a presence of one or more non-classical variant(s) of an amyloid beta precursor protein (APP) gene from a biological sample from an individual, comprising: - (a) detecting an expression profile or an activity profile of the one or more non-classical variant(s) of the APP gene in the biological sample from the individual by (i) long-read sequencing of RNA or DNA encoding for the one or more non-classical variant(s), or (ii) binding of one or more probe(s) to the one or more non-classical variant(s); and
- (b) identifying the presence of the one or more non-classical variant(s) when an elevated expression profile or elevated activity profile of the one or more non-classical variant(s) of an APP gene compared to a reference expression profile or reference activity profile of the one or more non-classical variant(s) derived from a cohort of control individuals is detected from step (a).
Provided herein, in some embodiments, are methods of notifying an individual of a presence of one or more non-classical variant(s) of an APP gene, comprising: - (a) obtaining an expression profile or an activity profile of the one or more non-classical variant(s) of the APP gene, wherein the expression profile or the activity profile is determined by (i) long-read sequencing of RNA or DNA encoding for the one or more non-classical variant(s), or (ii) binding of one or more probe(s) to the one or more non-classical variant(s); and
- (b) notifying an individual of the expression profile or the activity profile.
In some embodiments, the expression profile is expression level of the one or more non-classical variant(s), and wherein the activity profile is activity level of the one or more non-classical variant(s). In some embodiments, the expression profile is expression of a set of different non-classical variants, and wherein the activity profile is activity of a set of different non-classical variants. In some embodiments, the methods further comprise quantifying the expression level of the one or more non-classical variant(s) by a method comprising quantitative polymerase chain reaction (qPCR). In some embodiments, the one or more non-classical variant(s) comprises a portion or all ofexon exon 8. In some embodiments, one or more exon(s) in the one or more non-classical variant(s) is rearranged as compared to a control. In some embodiments, the one or more non-classical variant(s) comprises a single nucleotide variation (SNV) in APP. In some embodiments, the SNV in APP translates to amino acid positions in APP selected from a group consisting of A673V, A713T, T714I, V715M, V715A, I716M, V717I, V717F, T719P, and L723P. In some embodiments, the one or more non-classical variant(s) comprises a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16. In some embodiments, the disease or disorder is Alzheimer's disease. In some embodiments, Alzheimer's disease is familial Alzheimer's disease (FAD) or sporadic Alzheimer's disease. In some embodiments, the biological sample is blood or cerebrospinal fluid. In some embodiments, the biological sample comprises RNA, DNA, or protein encoding for the one or more non-classical variant(s). In some embodiments, the DNA is genomic DNA, extrachromosomal DNA, or circular DNA. In some embodiments, the one or more probe(s) are selected from polynucleotides or polypeptides. In some embodiments, the one or more probe(s) hybridize to RNA or DNA encoding for the one or more non-classical variant(s) within the biological sample. In some embodiments, the one or more probe(s) hybridize to a range of about 35 to about 50 nucleotides of the RNA or DNA. In some embodiments, the one or more probe(s) hybridize to an intraexonic junction betweenexon 1 andexon 11,exon 1 andexon 14,exon 2 andexon 17,exon 2 andexon 14,exon 2 andexon 18,exon 2 andexon 16,exon 3 andexon 16,exon 3 andexon 14,exon 3 andexon 17,exon 6 andexon 17,exon 6 andexon 18,exon 3 andexon 9,exon 2 andexon 9,exon 16 andexon 18,exon 6 andexon 12,exon 5 andexon 16, orexon 16 andexon 17. In some embodiments, methods further comprise capturing the one or more non-classical variant(s) from the biological sample prior to contacting the one or more non-classical variant(s) with the one or more probe(s). In some embodiments, the one or more non-classical variant(s) is detected by in situ hybridization or immunological hybridization. In some embodiments, the in situ hybridization is chromogenic in situ hybridization or fluorescence in situ hybridization. In some embodiments, binding of the one or more probe(s) to the one or more non-classical variant(s) further comprises a pull-down assay. In some embodiments, the one or more probe(s) for the pull-down assay are designed to hybridize to a portion or all ofexon
- Provided herein, in some embodiments, are kits for detecting one or more non-classical variant(s) of an APP gene in a biological sample of an individual, the kit comprising:
-
- (a) one or more probe(s) that hybridize to the one or more non-classical variant(s) of the APP gene or protein thereof; and
- (b) a detecting reagent for examining binding of the one or more probe(s) with the one or more non-classical variant(s) of the APP gene or protein thereof.
In some embodiments, the one or more non-classical variant(s) comprises a portion or all ofexon exon 8. In some embodiments, one or more exon(s) in the one or more non-classical variant(s) is rearranged as compared to a control. In some embodiments, the one or more non-classical variant(s) comprises a single nucleotide variation (SNV) in APP. In some embodiments, the SNV in APP translates to amino acid positions in APP selected from a group consisting of A673V, A713T, T714I, V715M, V715A, I716M, V717I, V717F, T719P, and L723P. In some embodiments, the one or more non-classical variant(s) comprises a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16. In some embodiments, the one or more probe(s) are selected from polynucleotides or polypeptides. In some embodiments, the one or more probe(s) hybridize to an intraexonic junction betweenexon 1 andexon 11,exon 1 andexon 14,exon 2 andexon 17,exon 2 andexon 14,exon 2 andexon 18,exon 2 andexon 16,exon 3 andexon 16,exon 3 andexon 14,exon 3 andexon 17,exon 6 andexon 17,exon 6 andexon 18,exon 3 andexon 9,exon 2 andexon 9,exon 16 andexon 18,exon 6 andexon 12,exon 5 andexon 16, orexon 16 andexon 17. In some embodiments, the one or more probe(s) bind to one or more protein(s) encoded by the one or more non-classical variant(s). In some embodiments, the one or more probe(s) comprise an antibody or fragment thereof. In some embodiments, a probe in the one or more probe(s) is labeled using a fluorochrome or a radioactive isotope. In some embodiments, a probe in the one or more probe(s) is labeled using an affinity tag. In some embodiments, the affinity tag is biotin, desthiobiotin, histidine, polyhistidine, myc, hemagglutinin (HA), FLAG, glutathione S transferase (GST), or derivatives thereof. In some embodiments, the detecting reagent binds to the one or more probe(s). In some embodiments, the detecting reagent comprises a fluorescent or a radioactive label. In some embodiments, the individual is suspected of having or being predisposed to Alzheimer's disease.
-
FIG. 1 illustrates a schema of amyloid precursor protein (APP) mRNA and cDNA. -
FIG. 2 illustrates a gel electrophoresis of RT-PCR from neurons isolated from cortices (CTX) and cerebellums (CBL) from non-diseased (Non-AD) postmortem brains and Alzheimer's disease (AD) brains. M represents DNA ladder marker. -
FIG. 3 illustrates a Southern blot of RT-PCR from neurons isolated from cortices (CTX) and cerebellums (CBL) from non-diseased (Non-AD) postmortem brains and Alzheimer's disease (AD) brains hybridized with APP cDNA probe. P and N represent positive (APP plasmid) and negative (Presenilin 1 plasmid) control, respectively. -
FIG. 4 illustrates a gel electrophoresis of RT-PCR from neurons isolated from cortices (CTX) and cerebellums (CBL) from non-diseased (Non-AD) postmortem brains and Alzheimer's disease (AD) brains. Arrows indicate positive signals corresponding to signal from Southern blot. -
FIG. 5 illustrates exemplary non-classical variants. -
FIG. 6A illustrates amyloid beta positive clones from non-diseased (Non-AD) brains and Alzheimer's disease (AD) brains detected with amyloid beta (Aβ) probes and APP cDNA probes. -
FIG. 6B illustrates a graph of percentage of amyloid beta in non-diseased (Non-AD) brains and Alzheimer's disease (AD) brains. -
FIG. 7 illustrates a gel electrophoresis of RT-PCR from frontal cortices of Alzheimer's disease (AD) brains (AD1 and AD3) in sorted neuronal and non-neuronal nuclei and bulk RNA. -
FIG. 8A illustrates a gel electrophoresis of PCR for genomic DNA isolated from frontal cortices of non-diseased brains and Alzheimer's disease (AD) brains (AD1 and AD3) with APP primers. NTC represents no template control. -
FIG. 8B illustrates a gel electrophoresis of PCR for genomic DNA isolated from frontal cortices of non-diseased brains and Alzheimer's disease (AD) brains (AD1 and AD3) with Presenilin 1 (PSEN1) primers. NTC represents no template control, and PC represents positive control. -
FIG. 9 illustrates a graph of relative ΔCT for neuronal and non-neuronal samples in which cAPP-R3/16 and PSEN are detected. -
FIG. 10 illustrates a schema for detection of non-classical variants by DNA pull-down assay. -
FIG. 11 illustrates genomic rearrangements in an Alzheimer's disease (AD) neuronal nuclei detected by DNA in situ hybridization. -
FIG. 12 illustrates intraexonic rearrangements in tissue sections from an Alzheimer's disease (AD) brain detected by RNA in situ hybridization of non-classical variant cAPP-R3/16 (red, 1201). Amyloid beta (Aβ) is stained in brown (1203). Tissue sections are visualized at 200× and 630× magnification. -
FIG. 13 illustrates non-classical variants detected by single molecule real-time sequencing. -
FIG. 14 illustrates the presence ofexon 16 andexon 17 junction in neuronal and non-neuronal nuclei from non-diseased and Alzheimer's disease (AD) brains detected by DNA in situ hybridization. -
FIG. 15A illustrates immunofluorescence images of cells transfected with non-classical variants cAPP-R3/16, cAPP-R2/18, and ncAPP-R1/4 that are hemagglutinin (HA) epitope tagged (left panel). EGFP co-expression (middle panel) and DAPI staining (right panel) are shown. -
FIG. 15B illustrates a Western blot of cell lysate from cells transfected with non-classical variants cAPP-R3/16, cAPP-R2/18, and ncAPP-R1/4 that are hemagglutinin (HA) epitope tagged. The Western blot was probed with HA antibody. -
FIG. 16A illustrates a gel electrophoresis of RT-PCR of LN-229 cells transfected with non-classical variant cAPP-R3/16 or wild-type APP (APP). -
FIG. 16B illustrates cAPP-R3/16 induction in LN-229 cells transfected with cAPP-R3/16 or wild-type APP (APP) detected by DNA in situ hybridization using cAPP-R3/16 probe and PPIB probe as a positive control. -
FIG. 17A illustrates a schematic for identification of non-classical RNA variants of APP from populations of neurons. (1) 50-neuronal nuclei were sorted from human prefrontal cortices (FCTX) by fluorescence-activated nuclear sorting (FANS) and used for (2) RT-PCR. Resulting RT-PCR products were screened by (3) Southern blot with 32P-labeled APP cDNA probes. (4) Bands with positive signals from duplicate gels were cloned and sequenced. (5) Non-classical variants were identified. Figure discloses SEQ ID NO: 33. -
FIG. 17B illustrates electrophoresis of RT-PCR products from 3 non-diseased and 3 sporadic AD brains. APP and PSEN1 plasmids were run as positive and negative controls for Southern blotting, respectively. -
FIG. 17C illustrates a gel electrophoresis of RT-PCR for bulk RNA detecting canonical APP splice variants as major products. Bulk RNA from 3 non-diseased and 3 sporadic Alzheimer's disease (SAD) prefrontal cortices was used for APP RT-PCR. Major products detected were canonical APP splice variants. -
FIG. 17D illustrates Southern blot of RT-PCR products. Arrows indicate examples of corresponding bands fromFIG. 17B that were cloned and Sanger sequenced. -
FIG. 17E illustrates a schema of structure of human APP genomic locus. -
FIG. 17F illustrates schemas of non-classical RNA variants of APP identified by RT-PCR. -
FIG. 17G illustrates schemas of sequence homology of non-classical RNA variants of APP at intraexonic junctions. Homology sequences of proximal and distal exons are shaded in gray. Middle sequence is the identified variant, top and bottom sequences are publicly available coding sequences from NM 000484.3 from the respective exons. Nucleotide variations are indicated an asterisk. Non-classical RNA variants identified by Sanger sequencing and PacBio data sets are shown with R and P, respectively. Figure discloses SEQ ID NOS 34-42, respectively, in order of appearance. -
FIG. 17H illustrates non-classical RNA variants of APP identified from independent long-read single molecule real-time (SMRT) sequencing data sets. -
FIG. 18A illustrates a schema for analyzing non-classical genomic cDNA (gencDNA) variants of APP. (1) Neuronal nuclei from human prefrontal cortices (FCTX) were used for (2) genomic DNA PCR and (3) junction-specific genomic in situ hybridization (jgISH). -
FIG. 18B illustrates a gel electrophoresis of genomic DNA PCR products with APP and PSEN1 primer sets using DNA from non-diseased (ND) and sporadic AD (SAD) neurons. Non-template control (NC) and positive control (PC) with indicated plasmids are shown. -
FIG. 18C illustrates a schema showing 13 variants identified first by RT-PCR (APP-R) and DNA PCR (APP-D). Seven were identified in both methods, five by RT-PCR only, and one by DNA PCR only. -
FIGS. 19A-19B illustrate jgISH performed with sense and anti-sense probes targetingAPP exon 16 andexon 17 junction (Ex 16/17) (FIG. 19A ) and intraexonic junction betweenAPP exon 3 and exon 16 (IEJ 3/16) on SAD neuronal nuclei (FIG. 19B ). -
FIGS. 19C-19D illustrate RNA-in situ hybridization with sense and antisense jgISH probes on human tissue sections.FIG. 19C showsEx 16/17 sense and anti-sense probes for RNA-in situ hybridization on human prefrontal cortex tissue sections.FIG. 19D showsIEJ 3/16 sense and anti-sense probes for RNA-in situ hybridization on human prefrontal cortex tissue sections. Scale bars are 10 μm. -
FIGS. 19E-19J illustrate restriction enzyme (RE) digestion. RE digestion was performed using MluCI (FIGS. 19E-19G ) and PstI+MslI (FIGS. 19H-19J ) to eliminateEx 16/17 andIEJ 3/16 target sequences, respectively.FIG. 19E andFIG. 19H illustrate jgISH nuclei.FIG. 19F andFIG. 19I illustrate graphs of quantification of average foci per nucleus; statistical significance was determined using the unpaired, two-tailed Mann-Whitney test.FIG. 19G andFIG. 19J illustrate cumulative frequency distributions represented as the number of foci per nucleus compared using the unpaired, two-tailed non-parametric Kolmogorov-Smirnov test. ****p<0.0001. n.s., not-significant. Error bars are ±SEM. Scale bars are 10 μm. -
FIG. 20A illustrates a schema of non-classical gencDNAs variants of APP identified by SMRT sequencing from SAD brains. (1) Neuronal nuclei from SAD prefrontal cortex (FCTX) were sorted and used for (2) genomic DNA PCR. Multiple reactions were pooled for (3) library preparation to enable (4) high fidelity sequencing (SMRT 20×CCS calling). -
FIG. 20B illustrates intraexonic junctions (IEJs). -
FIG. 20C illustrates a chart of total number and proportion of unique reads from each identified IEJ form. -
FIG. 20D illustrates a graph of number of unique IEJs forms. -
FIG. 20E illustrates a concentric circle plot of the APP locus (exon numbers along perimeter) illustrating IEJs (connecting lines inside the circles), deletions (DEL) (first inner circle), insertions (INS) (second inner circle), and single nucleotide variations (SNVs) (third inner circle) from sporadic Alzheimer's Disease (SAD) brains. Black dots indicate the abundance of DELs, INSs, and SNVs on a log(10) scale at the specified exon location. The outermost circle illustrates the sum count (key) of unique changes. Aβ region is highlighted, and known familial AD mutations are circled. -
FIG. 20F illustrates a concentric circle plot of the APP locus (exon numbers along perimeter) illustrating IEJs (connecting lines inside the circles), deletions (DEL) (first inner circle), insertions (INS) (second inner circle), and single nucleotide variations (SNVs) (third inner circle) from non-diseased brains. Black dots indicate the abundance of DELs, INSs, and SNVs on a log(10) scale at the specified exon location. Outermost circle illustrates the sum count (key) of unique changes. Aβ region is highlighted. -
FIG. 21A illustrates 10 different familial AD mutations present in APP gencDNAs. In-frame mutations (A673V, V715M, I716M, V717F, L723P, and V717I) out-of-frame mutations (V715A, T719P, A713T, T714I) are indicated based on the known APP reading frame analysis. -
FIGS. 21B-21C illustrate nuclei sorted from 6 ND and 6 SAD cortices. Nuclei were analyzed byEx 16/17 (FIG. 21B ) andIEJ 3/16 jgISH (FIG. 21C ). Cumulative frequency distribution plots of number of foci per nucleus showed statistical significance (nonparametric Kruskal-Wallis test with Dunn's multiple corrections). ****P<0.0001. n.s., not-significant. Error bars are ±SEM. Scale bars are 10 μm. -
FIGS. 21D-21I illustrate plots of relative percentage of nuclei sorted from 6 ND and 6 SAD cortices. Nuclei were analyzed byEx 16/17 (FIGS. 21D-21F ) andIEJ 3/16 (FIGS. 21G-21I ) jgISH. Cumulative frequency distribution plots of number of foci per nucleus showed statistical significance (nonparametric Kruskal-Wallis test with Dunn's multiple corrections). *p<0.05, **p<0.01, ***p<0.001, ****p<0.0001. n.s., not-significant. Error bars are ±SEM. -
FIG. 22A illustrates images ofIEJ 3/16 jgISH of nuclei isolated from the cortex of an AD mouse model (J20 transgenic, with neuron specific expression of human APP cDNA containing Swedish and Indiana mutations) versus WT littermates. Error bars are ±SEM. Scale bars are 10 μm. -
FIGS. 22B-22C illustrate quantification of nuclei fromFIG. 22A .FIG. 22B illustrates a cumulative frequency distribution illustrating the foci per nucleus.FIG. 22C illustrates a graph of an average number of foci per nucleus. ****p<0.0001. n.s., not-significant. -
FIGS. 22D-22G illustrateIEJ 3/16 jgISH from two mouse experiments.FIG. 22D illustrates a graph of foci per nucleus (x-axis) as relative percent of nuclei (y-axis) from J20+, WT+, J20−, and WT− from a first mouse experiment.FIG. 22E illustrates a graph of average foci per nucleus (y-axis) from J20+, WT+, J20−, and WT− mice from a first mouse experiment.FIG. 22F illustrates a graph of foci per nucleus (x-axis) as relative percent of nuclei (y-axis) from J20+, WT+, J20−, and WT− from a second mouse experiment.FIG. 22G illustrates a graph of average foci per nucleus (y-axis) from J20+, WT+, J20−, and WT− mice from a second mouse experiment. -
FIG. 23A illustrates images of representative nuclei isolated from 177 and 829-day J20 cortices. Scale bars are 10 μm. -
FIG. 23B illustrates a graph of cumulative percentage distribution of foci area (y-axis) as compared to relative foci area (x-axis) of nuclei fromFIG. 23A . -
FIG. 23C illustrates a graph of average foci area (y-axis) of nuclei fromFIG. 23A . -
FIG. 23D illustrates synthetic DNA targets containing theEx 16/17 junction sequence introduced by retroviral transduction in NIH-3T3 cells, and the target sequence (provirus) identified byEx 16/17 jgISH. -
FIGS. 23E-23F illustrates graphs of foci size from concatamer (×2) fromFIG. 23D .FIG. 23E illustrates a graph of cumulative percentage of foci (y-axis) as compared to relative foci area (x-axis).FIG. 23F illustrates average foci area (x-axis) from x0, x1, x2 ofFIG. 23D . ****p<0.0001. n.s., not-significant. Error bars are ±SEM. -
FIG. 24 illustrates cortical tissue sections from human Alzheimer's disease subjects stained with antibodies recognizing AP peptide (“Aβ”) and APP c-terminal (“c-terminal”). “Merge” images illustrate co-localization of AP peptide with APP c-terminal. “DAPI” images illustrate nuclei staining. - Throughout this disclosure, various embodiments are presented in a range format. It should be understood that the description in range format is merely for convenience and brevity and should not be construed as an inflexible limitation on the scope of any embodiments. Accordingly, the description of a range should be considered to have specifically disclosed all the possible subranges as well as individual numerical values within that range to the tenth of the unit of the lower limit unless the context clearly dictates otherwise. For example, description of a range such as from 1 to 6 should be considered to have specifically disclosed subranges such as from 1 to 3, from 1 to 4, from 1 to 5, from 2 to 4, from 2 to 6, from 3 to 6 etc., as well as individual values within that range, for example, 1.1, 2, 2.3, 5, and 5.9. This applies regardless of the breadth of the range. The upper and lower limits of these intervening ranges may independently be included in the smaller ranges, and are also encompassed within the invention, subject to any specifically excluded limit in the stated range. Where the stated range includes one or both of the limits, ranges excluding either or both of those included limits are also included in the invention, unless the context clearly dictates otherwise.
- The terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting of any embodiment. As used herein, the singular forms “a,” “an” and “the” are intended to include the plural forms as well, unless the context clearly indicates otherwise. It will be further understood that the terms “comprises” and/or “comprising,” when used in this specification, specify the presence of stated features, integers, steps, operations, elements, and/or components, but do not preclude the presence or addition of one or more other features, integers, steps, operations, elements, components, and/or groups thereof. As used herein, the term “and/or” includes any and all combinations of one or more of the associated listed items.
- Unless specifically stated or obvious from context, as used herein, the term “about” in reference to a number or range of numbers is understood to mean the stated number and numbers+/−10% thereof, or 10% below the lower listed limit and 10% above the higher listed limit for the values listed for a range.
- The term “non-classical variant” as used herein refers to RNA or DNA molecules comprising intraexonic junctions between exons and/or conventional spliced exon-exon junctions, and RNA or DNA molecules lacking introns between exons.
- The term “genomic cDNA” or “gencDNA” as used herein refers to a genomic variant lacking introns. In some embodiments, the gencDNA comprises intraexonic junctions between exons. In some embodiments, the gencDNA comprises inverted exons. In some embodiments, the gencDNA is generated by reverse transcription of a non-classical RNA variant. In some embodiments, the gencDNA is incorporated into genomic DNA.
- The term “expression” as used herein refers to a transcriptional or translational product of a gene.
- The term “activity” as used herein refers to protein biological or chemical function.
- The term “RNA” as used herein refers to a molecule comprising at least one ribonucleotide residue. By “ribonucleotide” is meant a nucleotide with a hydroxyl group at the 2′ position of a beta-D-ribo-furanose moiety. The term RNA includes, but not limited to, mRNA, ribosomal RNA, tRNA, non-protein-coding RNA (npcRNA), non-messenger RNA, functional RNA (fRNA), long non-coding RNA (lncRNA), pre-mRNAs, and primary miRNAs (pri-miRNAs). The term RNA includes, for example, double-stranded (ds) RNAs; single-stranded RNAs; and isolated RNAs such as partially purified RNA, essentially pure RNA, synthetic RNA, recombinantly produced RNA, as well as altered RNA that differ from naturally-occurring RNA by the addition, deletion, substitution and/or alteration of one or more nucleotides. Such alterations can include addition of non-nucleotide material, such as to the end(s) of the siRNA or internally, for example at one or more nucleotides of the RNA. Nucleotides in the RNA molecules described herein can also comprise non-standard nucleotides, such as non-naturally occurring nucleotides or chemically synthesized nucleotides or deoxynucleotides. These altered RNAs can be referred to as analogs or analogs of naturally-occurring RNA.
- The term “RNAi” as used herein refers to an RNA molecule that induces RNA interference (RNAi). In some embodiments, the RNAi molecule is a dsRNA molecule that will generate a siRNA molecule or miRNA molecule following contact with Dicer (i.e., an RNAi molecule precursor). In some embodiments, the RNAi molecule is a siRNA duplex, a siRNA sense molecule, a siRNA anti-sense molecule, a miRNA duplex, a miRNA sense molecule, a miRNA anti-sense molecule, and analogues thereof.
- The terms “binding fragment,” “antibody fragment,” or “antigen binding fragment” are used herein, for purposes of the specification and claims, to mean a portion or fragment of an intact antibody molecule, preferably wherein the fragment retains antigen-binding function. Examples of antibody fragments include Fab, Fab′, F(ab′)2, Fd, Fd′ and Fv fragments, diabodies, linear antibodies, single-chain antibody molecules, single-chain binding polypeptides, scFv, bivalent scFv, tetravalent scFv, and bispecific or multispecific antibodies formed from antibody fragments.
- “Fab” fragments are typically produced by papain digestion of antibodies resulting in the production of two identical antigen-binding fragments, each with a single antigen-binding site and a residual “Fc” fragment. Pepsin treatment yields a F(ab′)2 fragment that has two antigen-combining sites capable of cross-linking antigen. An “Fv” is the minimum antibody fragment that contains a complete antigen recognition and binding site. In a two-chain Fv species, this region consists of a dimer of one heavy- and one light-chain variable domain in tight, non-covalent association. In a single-chain Fv (scFv) species, one heavy- and one light-chain variable domain are covalently linked by a flexible peptide linker such that the light and heavy chains associate in a “dimeric” structure analogous to that in a two-chain Fv species. It is in this configuration that the three CDRs of each variable domain interact to define an antigen-binding site on the surface of the VH-VL dimer. Collectively, the six CDRs confer antigen-binding specificity to the antibody. However, even a single variable domain (or half of an Fv comprising only three CDRs specific for an antigen) has the ability to recognize and bind antigen, although usually at a lower affinity than the entire binding site.
- The Fab fragment also contains the constant domain of the light chain and the first constant domain (CH1) of the heavy chain. Fab fragments differ from Fab′ fragments by the addition of a few residues at the carboxy terminus of the heavy-
chain C H1 domain including one or more cysteines from the antibody hinge region. Fab′-SH is the designation herein for Fab′ in which the cysteine residue(s) of the constant domains bear a free thiol group. F(ab′)2 antibody fragments originally were produced as pairs of Fab′ fragments that have hinge cysteines between them. Other chemical couplings of antibody fragments are also suitable. - The term “monoclonal antibody” refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations that are present in minor amounts. In some embodiments, monoclonal antibodies are made, for example, by the hybridoma method. In some embodiments, monoclonal antibodies are isolated from phage antibody libraries.
- The antibodies herein include monoclonal, polyclonal, recombinant, chimeric, humanized, bi-specific, grafted, human, and fragments thereof including antibodies altered by any means to be less immunogenic in humans. Thus, for example, the monoclonal antibodies and fragments herein include “chimeric” antibodies and “humanized” antibodies. In general, chimeric antibodies include a portion of the heavy and/or light chain that is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, so long as they exhibit the desired biological activity. For example in some embodiments, a chimeric antibody contains variable regions derived from a mouse and constant regions derived from human in which the constant region contains sequences homologous to both human IgG2 and human IgG4. Numerous methods for preparing “chimeric” antibodies are known in the art. “Humanized” forms of non-human (e.g., murine) antibodies or fragments are chimeric immunoglobulins, immunoglobulin chains or fragments thereof (such as Fv, Fab, Fab′, F(ab′)2 or other antigen-binding subsequences of antibodies) which contain minimal sequence derived from non-human immunoglobulin. Humanized antibodies include, grafted antibodies or CDR grafted antibodies wherein part or all of the amino acid sequence of one or more complementarity determining regions (CDRs) derived from a non-human animal antibody is grafted to an appropriate position of a human antibody while maintaining the desired binding specificity and/or affinity of the original non-human antibody. In some embodiments, corresponding non-human residues replace Fv framework residues of the human immunoglobulin. In some embodiments, humanized antibodies comprise residues that are found neither in the recipient antibody nor in the imported CDR or framework sequences. These modifications are made to further refine and optimize antibody performance. In some embodiments, the humanized antibody comprises substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin consensus sequence. Numerous methods for “humanizing” antibodies are known in the art.
- Alzheimer's disease (AD) is a chronic neurodegenerative disease resulting in deterioration of cognitive function. AD is a common form of dementia and estimated to cause 60-70% of the ˜48 million people worldwide with dementia. AD can be classified as familial Alzheimer's disease (FAD), which comprises a genetic component, and sporadic AD (SAD) that can arise from several factors including genetic factors.
- AD neuropathology is characterized by accumulation of amyloid beta protein and neurofibrillary tangles comprising Tau in the Central Nervous System, synaptic loss, and neuronal death. Specifically, accumulation of amyloid beta as amyloid beta protein plaques or soluble amyloid beta oligomers has been implicated in AD progression.
- Amyloid beta protein results from cleavage of amyloid beta precursor protein (APP) by α-, γ-, and β-secretases. In some instances, cleavage of APP is amyloidogenic and results in unwanted accumulation of amyloid beta protein.
- Antibody therapies for treating Alzheimer's disease are primarily aimed at targeting unwanted accumulation of amyloid beta protein. As a result, unwanted amyloid beta protein is cleared from the brain. These treatments, however, result in only partial success. Thus, successful treatments for Alzheimer's disease need to account for the genetic complexity of the disease.
- Amyloid precursor protein (APP) gene copy number has been implicated in AD pathogenesis. For example, Down Syndrome (DS) where there are three copies of APP results in neuropathology similar to AD. In addition, increased APP gene copy number has been observed in AD brains. In some instances, the increased gene copy number is a result of genomic rearrangements such as intraexonic rearrangements that generate non-classical variants of APP.
- Described herein are non-classical variants of APP gene. Non-classical variants as described herein comprise, in some embodiments, intraexonic rearrangements. In some embodiments, the non-classical variants lack introns. In some embodiments, the non-classical variants comprise a portion of a first exon of APP and a portion of a second exon of APP. In some embodiments, the non-classical variants comprise intraexonic junctions and lack introns. In some embodiments, the non-classical variants comprise inverted exons. In some embodiments, the non-classical variants are genomic cDNAs (gencDNAs). In some embodiments, the non-classical variants comprise a single nucleotide variant (SNV). In some embodiments, non-classical variants comprise deletion of exons. In some embodiments, non-classical variants comprise insertions. In some embodiments, non-classical variants comprise copy number variation (CNV), L1 repeat elements, SNVs, deletions, insertions, intraexonic junctions, or combinations thereof. In some embodiments, the SNVs are somatic SNVs. In some embodiments, the SNVs are germline SNVs.
- Various mechanisms for generation of non-classical variants of APP are contemplated herein. In some embodiments, the non-classical variants of APP are generated by DNA damages induced DNA repair. In some embodiments, the non-classical variants of APP are generated by a homologous recombination event. In some embodiments, the non-classical variants of APP are generated by a non-homologous recombination event. In some embodiments, a reverse transcriptase is involved in generating the non-classical variants of APP. In some embodiments, a non-classical variant of APP is generated by activity of a DNA polymerase. In some embodiments, generation of the non-classical variants of APP comprises a RNA splicing event. Generation of a non-classical variant of APP, in certain embodiments, involves transcription. In some embodiments, generation of the one or more non-classical variant(s) involves reverse transcription. In some embodiments, the transcription is cell-type specific. For example, the non-classical variants are generated by neuron-specific RNA transcription.
- In some embodiments, a non-classical a variant of APP is generated by incorporation of RNA intermediates into genomic DNA. In some embodiments, the RNA intermediates are non-classical RNA variants of APP. In some embodiments, the RNA intermediates are reversed transcribed and introduced into the genomic DNA. In some embodiments, the RNA intermediates that are reversed transcribed are gencDNAs. In some embodiments, incorporation of RNA intermediates into the genomic DNA involves a break in the DNA. In some embodiments, the break is a single-stranded break. In some embodiments, the break is a double-stranded break. In some embodiments, the break is introduced by an enzyme, a chemical, or radiation.
- In some embodiments, generation of a non-classical variant of APP involves alteration in a DNA repair pathway. Exemplary DNA repair pathways include, but are not limited to, non-homologous end joining (NHEJ), microhomology-mediated end joining (MMEJ), homologous recombination, mismatch repair, nucleotide excision repair, or DNA strand cross-link repair.
- In some embodiments, generation of a non-classical variant of APP involves RNA processing. In some embodiments, generation of the non-classical variants of APP comprises a RNA splicing event. In some embodiments, generation of the non-classical variants of APP comprises a RNA alternative splicing event. Exemplary alternative splicing events include, but are not limited, intron retention, exon skipping, alternative 5′ splice site, alternative 3′ splice site, and mutually exclusive exons.
- In some embodiments, the non-classical variants are coding. In some embodiments, the non-classical variants are non-coding. In some embodiments, the non-classical variants comprise RNA or DNA. In some embodiments, the non-classical variants comprise genomic cDNA (gencDNA).
- In some embodiments, the non-classical variants comprise a portion or all of an exon of APP. In some embodiments, the non-classical variants comprise a portion or all of
exon exon exon 8. In some embodiments, the non-classical variants do not compriseexon 7. In some embodiments, the non-classical variants do not compriseexon 8 andexon 7. In some embodiments, the non-classical variants of APP comprise a portion or all ofexon exon - For example, the non-classical variants comprise nucleotides 1-24 of
exon 3 of APP and nucleotides 45-101 ofexon 16 of APP. In some embodiments, the non-classical variants comprise nucleotides from more than one exon of APP. In some embodiments, the non-classical variants comprise nucleotides from at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, or 17 exons of APP. In some embodiments, the non-classical variants comprise a portion of at most 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, or 17 exons of APP. - In some embodiments, non-classical variants of APP comprise a portion of
exon 1 and a portion ofexon exon 1 and at least or about 10 nt, 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt ofexon - In some embodiments, non-classical variants of APP comprise a portion of
exon 2 and a portion ofexon exon 2 and at least or about 10 nt, 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt ofexon - In some embodiments, non-classical variants of APP comprise a portion of
exon 3 and a portion ofexon exon 3 and at least or about 10 nt, 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt ofexon - In some embodiments, non-classical variants of APP comprise a portion of
exon 4 and a portion ofexon exon 4 and at least or about 10 nt, 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt ofexon - In some embodiments, non-classical variants of APP comprise a portion of
exon 5 and a portion ofexon exon 5 and at least or about 10 nt, 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt ofexon - In some embodiments, non-classical variants of APP comprise a portion of
exon 6 and a portion ofexon exon 6 and at least or about 10 nt, 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt ofexon - In some embodiments, non-classical variants of APP comprise a portion of
exon 7 and a portion ofexon exon 7 and at least or about 10 nt, 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt ofexon - In some embodiments, non-classical variants of APP comprise a portion of
exon 8 and a portion ofexon exon 8 and at least or about 10 nt, 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt ofexon - In some embodiments, non-classical variants of APP comprise a portion of
exon 9 and a portion ofexon exon 9 and at least or about 10 nt, 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt ofexon - In some embodiments, non-classical variants of APP comprise a portion of
exon 10 and a portion ofexon exon 10 and at least or about 10 nt, 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt ofexon - In some embodiments, non-classical variants of APP comprise a portion of
exon 11 and a portion ofexon exon 11 and at least or about 10 nt, 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt ofexon - In some embodiments, non-classical variants of APP comprise a portion of
exon 12 and a portion ofexon exon 12 and at least or about 10 nt, 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt ofexon - In some embodiments, non-classical variants of APP comprise a portion of
exon 13 and a portion ofexon exon 13 and at least or about 10 nt, 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt ofexon - In some embodiments, non-classical variants of APP comprise a portion of
exon 14 and a portion ofexon exon 14 and at least or about 10 nt, 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt ofexon - In some embodiments, non-classical variants of APP comprise a portion of
exon 15 and a portion ofexon exon 15 and at least or about 10 nt, 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt ofexon - In some embodiments, non-classical variants of APP comprise a portion of
exon 16 and a portion ofexon exon 16 and at least or about 10 nt, 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt ofexon - In some embodiments, non-classical variants of APP comprise a portion of
exon 17 and a portion ofexon exon 17 and at least or about 10 nt, 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt ofexon - In some embodiments, non-classical variants of APP comprise a portion of
exon 18 and a portion ofexon exon 18 and at least or about 10 nt, 15 nt, 20 nt, 25 nt, 30 nt, 40 nt, 50 nt, 75 nt, 100 nt, 150 nt, 200 nt, or more than 200 nt ofexon - Non-classical transcript variants as described herein, in certain embodiments, comprise portions of at least 2 exons. In some embodiments, the non-classical variants comprise a portion of
exon 1 and a portion ofexon 11. In some embodiments, the non-classical variants comprise a portion ofexon 1 and a portion ofexon 12. In some embodiments, the non-classical variants comprise a portion ofexon 1 and a portion ofexon 14. In some embodiments, the non-classical variants comprise a portion ofexon 1 and a portion ofexon 17. In some embodiments, the non-classical variants comprise a portion ofexon 2 and a portion ofexon 14. In some embodiments, the non-classical variants comprise a portion ofexon 2 and a portion ofexon 16. In some embodiments, the non-classical variants comprise a portion ofexon 2 and a portion ofexon 17. In some embodiments, the non-classical variants comprise a portion ofexon 2 and a portion ofexon 18. In some embodiments, the non-classical variants comprise a portion ofexon 3 and a portion ofexon 9. In some embodiments, the non-classical variants comprise a portion ofexon 3 and a portion ofexon 14. In some embodiments, the non-classical variants comprise a portion ofexon 3 and a portion ofexon 16. In some embodiments, the non-classical variants comprise a portion ofexon 3 and a portion ofexon 17. In some embodiments, the non-classical variants comprise a portion ofexon 3 and a portion ofexon 18. In some embodiments, the non-classical variants comprise a portion ofexon 5 and a portion ofexon 16. In some embodiments, the non-classical variants comprise a portion of exon compriseexon 6 and a portion ofexon 12. In some embodiments, the non-classical variants comprise a portion ofexon 6 and a portion ofexon 16. In some embodiments, the non-classical variants comprise a portion ofexon 6 and a portion ofexon 17. In some embodiments, the non-classical variants comprise a portion ofexon 6 and a portion ofexon 18. In some embodiments, the non-classical variants comprise a portion ofexon 16 and aportion exon 18. - Described herein, in certain embodiments, are non-classical variants of APP comprising portions of at least two exons, wherein the at least two exons are linked by intraexonic junctions. In some embodiments, the intraexonic junction is between
exon 1 andexon exon 2 andexon exon 3 andexon exon 4 andexon exon 5 andexon exon 6 andexon exon 7 andexon exon 8 andexon exon 9 andexon exon 10 andexon exon 11 andexon exon 12 andexon exon 13 andexon exon 14 andexon exon 15 andexon exon 16 andexon exon 17 andexon exon 18 andexon - In some embodiments, sequence complementarity exists in non-classical variants of APP in the intraexonic junctions. In some embodiments, the sequence complementarity is at least or about 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 50, 60, 70, 80, 90, 100, or more than 100 nucleotides. In some embodiments, the sequence complementarity is in a range of about 2 nucleotides to 20 nucleotides. In some embodiments, the sequence complementarity is in a range of about 2 nucleotides to about 200 nucleotides, about 4 nucleotides to about 180 nucleotides, about 6 nucleotides to about 160 nucleotides, about 8 nucleotides to about 140 nucleotides, about 10 nucleotides to about 120 nucleotides, about 12 nucleotides to about 100 nucleotides, about 14 nucleotides to about 80 nucleotides, about 16 nucleotides to about 60 nucleotides, or about 20 nucleotides to about 40 nucleotides.
- Non-classical variants of APP as described herein, in certain embodiments, comprise different sequences. In some embodiments, a number of different sequences is at least or about 2, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, or more than 1000 sequences. In some embodiments, a number of different sequences in a range of about 2 sequences to about 1000 sequences, about 4 sequences to about 900 sequences, about 6 sequences to about 800 sequences, about 8 sequences to about 700 sequences, about 10 sequences to about 600 sequences, about 20 sequences to about 500 sequences, about 30 sequences to about 400 sequences, about 40 sequences to about 300 sequences, about 50 sequences to about 200 sequences, and about 60 sequences to about 100 sequences.
- In some embodiments, non-classical variants of APP comprising portions of at least 2 exons further comprise a deletion of at least one exon or a portion of at least one exon of APP. For example, the non-classical transcript variants comprise a deletion of
exon 8 or a portion ofexon 8 of APP. In some embodiments, the non-classical transcript variants comprise a deletion ofexon 7 or a portion ofexon 7 of APP. In some embodiments, the non-classical transcript variants comprise a deletion of exon or a portion ofexon exon - The APP mRNA sequence is set forth at NCBI Reference Sequence: NM_000484.3. Exemplary non-classical variants are illustrated in Table 1.
-
TABLE 1 SEQ ID NO Name* Sequence** 1 cAPP- ATGCTGCCCGGTTTGGCACTGCTCCTGCTGGCCGCCTGGACGGCTC R3/16 GGGCGCTGGAGGTACCCACTGATGGTAATGCTGGCCTGCTGGCTG AACCCCAGATTGCCATGTTCTGTGGCAGACTGAACATGCACATGA ATGTCCAGAATGGGAAGTGGGATTCAGATCCATCAGGGACCAAAA CCTGCATTGATACCAAGGAAGGCATCCTGCAGTATTGCCAAGAAG TCTACCCTGAACTGCAGATCAC C:A AGATGGATGCAGAATTCCGAC ATGACTCAGGATATGAAGTTCATCATCAAAAATTGGTGTTCTTTGC AGAAGATGTGGGTTCAAACAAAGGTGCAATCATTGGACTCATGGT GGGCGGTGTTGTCATAGCGACAGTGATCGTCATCACCTTGGTGATG CTGAAGAAGAAACAGTACACATCCATTCATCATGGTGTGGTGGAG GTTGACGCCGCTGTCACCCCAGAGGAGCGCCACCTGTCCAAGATG CAGCAGAACGGCTACGAAAATCCAACCTACAAGTTCTTTGAGCAG ATGCAGAACTAG 2 cAPP- ATGCTGCCCGGTTTGGCACTGCTCCTGCTGGCCGCCTGGACGGCTC R3/16-2 GGGCGCTGGAGGTACCCACTGATGGTAATGCTGGCCTGCTGGCTG AACCCCAGATTGCCATGTTCTGTGGCAGACTGAACATGCACATGA ATGTCCAGAATGGGAAGTGGGATTCAGATCCATCAGGGACCAAAA CCTGCATTGATACCAAGGAAGGCATCCTGCAGTATTGCCAAGAAG TCTACCCTGAACTGCAGATCAC C:A AGATGGATGCAGAATTCCGAC ATGACTCAGGATATGAAGTTCATCATCAAAAATTGGTGTTCTTTGC AGAAGATGTGGGTTCAAACAAAGGTGCAATCATTGGACTCATAGT GGGCGGTGTTGTCATAGCGACAGTGATCGTCATCACCTTGGTGATG CTGAAGAAGAAACAGTACACATCCATTCATCATGGTGTGGTGGAG GTTGACGCCGCTGTCACCCCAGAGGAGCGCCACCTGTCCAAGATG CAGCAGAACGGCTACGAAAATCCAACCTACAAGTTCTTTGAGCAG ATGCAGAACTAG 3 cAPP- ATGCTGCCCGGTTTGGCACTGCTCCTGCTGGCCGCCTGGACGGCTC R2/18 GGGCGCTGGAGGTACCCACTGATGGTAATGCTGGCCTGCTGGCTG AACCCCAGATTGCCATGTTCTGTGGCAGACTGAACATGCACATGA ATGTCCAGAATGGGAAGTGGGATTCAGATCCATCAGGGACCAAAA CCTGCATTGATACCAAGGAAGGCATCCT G:C GCTACGAAAATCCAA CCTACAAGTTCTTTGAGCAGATGCAGAACTAG 4 cAPP- ATGCTGCCCGGTTTGGCACTGCTCCTGCTGGCCGCCTGGACGGCTC R6/18 GGGCGCTGGAGGTACCCACTGATGGTAATGCTGGCCTGCTGGCTG AACCCCAGATTGCCATGTTCTGTGGCAGACTGAACATGCACATGA ATGTCCAGAATGGGAAGTGGGATTCAGATCCATCAGGGACCAAAA CCTGCATTGATACCAAGGAAGGCATCCTGCAGTATTGCCAAGAAG TCTACCCTGAACTGCAGATCACCAATGTGGTAGAAGCCAACCAAC CAGTGACCATCCAGAACTGGTGCAAGCGGGGCCGCAAGCAGTGCA AGACCCATCCCCACTTTGTGATTCCCTACCGCTGCTTAGTTGGTGA GTTTGTAAGTGATGCCCTTCTCGTTCCTGACAAGTGCAAATTCTTA CACCAGGAGAGGATGGATGTTTGCGAAACTCATCTTCACTGGCAC ACCGTCGCCAAAGAGACATGCAGTGAGAAGAGTACCAACTTGCAT GACTACGGCATGTTGCTGCCCTGCGGAATTGACAAGTTCCGAGGG GTAGAGTTTGTGTGTTGCCCACTGGCTGAAGAAAGTGACAATGTG GATTCTGCTGATGCGGAGGAGGATGACTCGGATGTCTGGTGGGGC GGAGCAGACACAGACTATGCAGATGGGAGTGAAGACAAAGTAGT AGAAGTAGCAGAGGAGGAAGAAGTGGCTGAGGTGGAAGAAGAAG AAGCCGATGATGA C:G AGGAGCGCCACCTGTCCAAGATGCAGCAG AGCGGCTACGAAAATCCAACCTACAAGTTCTTTGAGCAGATGCAG AACTAG 5 cAPP- ATGCTGCCCGGTTTGGCACTGCTCCTGCTGGCCGCCTGGACGGCTC R3/14 GGGCGCTGGAGGTACCCACTGATGGTAATGCTGGCCTGCTGGCTG AACCCCAGATTGCCATGTTCTGTGGCAGACTGAACATGCACATGA ATGTCCAGAATGGGAAGTGGGATTCAGATCCATCAGGGACCAAAA CCTGCATTGATACCAAGGAAGGCATCCTGCAGTATTGCCAAGAAG TCTACCCTGAACTGCAGATCACCAATGTGGTAGAAGCCAA C:A CAG AAAACGAAGTTGAGCCTGTTGATGCCCGCCCTGCTGCCGACCGAG GACTGACCACTCGACCAGGTTCTGGGTTGACAAATATCAAGACGG AGGAGATCTCTGAAGTGAAGATGGATGCAGAATTCCGACATGACT CAGGATATGAAGTTCATCATCAAAAATTGGTGTTCTTTACAGAAGA TGTGGGTTCAAACAAAGGTGCAATCATTGGACTCATGGTGGGCGG TGTTGTCATAGCGACAGTGATCGTCATCACCTTGGTGATGCTGAAG AAGAAACAGTACACATCCATTCATCATGGTGTGGTGGAGGTTGAC GCCGCTGTCACCCCAGAGGAGCGCCACCTGTCCAAGATGCAGCAG AACGGCTACGAAAATCCAACCTACAAGTTCTTTGAGCAGATGCAG AACTAG 6 ncAPP- ATGCTGCCCGGTTTGGCACTGCTCCTGCTGGCCGCCTGGACGGCTC R3/17 GGGCGCTGGAGATACCCACTGATGGTAATGCTGGCCTGCTGGCTG AACCCCAGATTGCCATGTTCTGTGGCAGACTGAACATGCACATGA ATGTCCAGAATGGGAAGTGGGACTCAGATCCATCAGGGACCAAAA CCTGCATTGATACCAAGGAAGGCATCCTGCAGTATTGCCAAGAAG TCTACCCTGAACTGCAGATCACCAATGTGGTAGAAGCCAACCAAC CAGTGACCATCCAGAACTGGTGCAAGCGGGGCCGCAAGCA G:T GT TGTCATAGCGACAGTGATCGTCATCACCTTGGTGATGCTGAAGAA GAAACAGTACACATCCATTCATCATGGTGTGGTGGAGGTTGACGC CGCTGTCACCCCAGAGGAGCGCCACCTGTCCAAGATGCAGCAGAA CGGCTACGAAAATCCAACCTACAAGTTCTTTGAGCAGATGCAGAA CTAG 7 cAPP- ATGCTGCCCGGTTTGGCACTGCTCCTGCTGGCCGCCTGGAC G:G CC R1/11 TCGTCACGTGTTCAATATGCTAAAGAAGTATGTCCGCGCAGAACA GAAGGACAGACAGCACACCCTAAAGCATTTCGAGCATGTGCGCAT GGTGGATCCCAAGAAAGCCGCTCAGATCCGGTCCCAGGTTATGAC ACACCTCCGTGTGATTTATGAGCGCATGAATCAGTCTCTCTCCCTG CTCTACAACGTGCCTGCAGTGGCCGAGGAGATTCAGGATGAAGTT GATGAGCTGCTTCAGAAAGAGCAAAACTATTCAGATGACGTCTTG GCCAACATGATTAGTGAACCAAGGATCAGTTACGGAAACGATGCT CTCATGCCATCTTTGACCGAAACGAAAACCACCGTGGAGCTCCTTC CCGTGAATGGAGAGTTCAGCCTGGACGATCTCCAGCCGTGGCATT CTTTTGGGGCTGACTCTGTGCCAGCCAACACAGAAAACGAAGTTG AGCCTGTTGATGCCCGCCCTGCTGCCGACCGAGGACTGACCACTCG ACCAGGTTCTGGGTTGACAAATATCAAGACGGAGGAGATCTCTGA AGTGAAGATGGATGCAGAATTCCGACATGACTCAGGATATGAAGT TCATCATCAAAAATTGGTGTTCTTTGCAGAAGATGTGGGTTCAAAC AAAGGTGCAATCATTGGACTCATGGTGGGTGGTGTTGTCATAGCG ACAGTGATCGTCATCACCTTGGTGATGCTGAAGAAGAAACAGTAC ACATCCATTCATCATGGTGTGGTGGAGGTTGACGCCGCTGTCACCC CAGAGGAGCGCCACCTGTCCAAGATGCAGCAGAACGGCTACGAAA ATCCAACCTACAAGTTCTTTGAGCAGATGCAGAACTAG 8 ncAPP- ATGCTGCCCGGTTTGGCACTGCTCCTGCAGTG G:G AGGAGATTCAG R1/13 GATGAAGTTGATGAACTGCTTCAGAAAGAGCAAAACTATTCAGAT GACGTCTTGGCCAACATGATTAGTGAACCAAGGATCAGTTACGGA AACGATGCTCTCATGCCATCTTTGACCGAAACGAAAACCACCGTG GAGCTCCTTCCCGTGAATGGAGAGTTCAGCCTGGACGATCTCCAGC CGTGGCATTCTTTTGGGGCTGACTCTGTGCCAGCCAACACAGAAAA CGAAGTTGAGCCTGTTGATGCCCGCCCTGCTGCCGACCGAGGACT GACCACTCGACCAGGTTCTGGGTTGACAAATATCAAGACGGAGGA GATCTCTGAAGTGAAGATGGATGCAGAATTCCGACATGACTCAGG ATATGAAGTTCATCATCAAAAATTGGTGTTCTTTGCAGAAGATGTG GGTTCAAACAAAGGTGCAATCATTGGACTCATGGTGGCGGTGTTG TCATAGCGACAGTGATCGTCATCACCTTGGTGATGCTGAAGAAGA AACAGTACACATCCATTCATCATGGTGTGGTGGAGGTTGACGCCG CTGTCACCCCAGAGGAGCGCCACCTGTCCAAGATGTGGCAGAACG GCTACGAAAATCCAACCTACAAGTTCTTTGAGCAGATGCAGAACT AG 9 ncAPP- ATGCTGCCCGGTTTGGCACTGCT C:T GCAGGCTGTTCCTCCTCGGCC R1/11-2 TCGTCACGTGTTCAATATGCTAAAGAAGTATGTCCGCGCAGAACA GAAGGACAGACAGCACACCCTAAAGCATTTCGAGCATGTGCGCAT GGTGGATCCCAAGAAAGCCGCTCAGATCCGGTCCCAGGTTATGAC ACACCTCCGTGTGATTTATGAGCGCATGAATCAGTCTCTCTCCCTG CTCTACAACGTGCCTGCAGTGGCCGAGGAGATTCAGGATGAAGTT GATGAGCTGCTTCAGAAAGAGCAAAACTATTCAGATGACGTCTTG GCCAACATGATTAGTGAACCAAGGATCAGTTACGGAAACGATGCT CTCATGCCATCTTTGACCGAAACGAAAACCACCGTGGAGCTCCTTC CCGTGAATGGAGAGTTCAGCCTGGACGATCTCCAGCCGTGGCATT CTTTTGGGGCTGACTCTGTGCCAGCCAACACAGAAAACGAAGTTG AGCCTGTTGATGCCCGCCCTGCTGCCGACCGAGGACTGACCACTCG ACCAGGTTCTGGGTTGACAAATATCAAGACGGAGGAGATCTCTGA AGTGAAGATGGATGCAGAATTCCGACATGACTCAGGATATGAAGT TCATCATCAAAAATTGGTGTTCTTTGCAGAAGATGTGGGTTCAAAC AAAGGTGCAATCATTGGACTCATGGTGGGCGGTGTTGTCATAGCG ACAGTGATCGTCATCACCTTGGTGATGCTGAAGAAGAAACAGTAC ACATCCATTCATCATGGTGTGGTGGAGGTTGACGCCGCTGTCACCC CAGAGGAGCGCCACCTGTCCAAGATGCAGCAGAACGGCTACGAAA ATCCAACCTACAAGTTCTTTGAGCAGATGCAGAACTAG 10 ncAPP- ATGCTGCCCGGTTTGGCACTGCTCCTGCTGGCCGCCTGGACAGC T: R1/14 C CTTCCCGTGAATGGAGAGTTCAGCCTGGACGATCTCCAGCCGTG GCATTCTTTTGGGGCTGACTCTGTGCCAGCCAACACAGAAAACGA AGTTGAGCCTGTTGATGCCCGCCCTGCTGCCGACCGAGGACTGACC ACTCGACCAGGTTCTGGGTTGACAAATATCAAGACGGAGGAGATC TCTGAAGTGAAGATGGATGCAGAATTCCGACATGACTCAGGATAT GAAGTTCATCATCAAAAATTGGTGTTCTTTGCAGAAGATGTGGGTT CAAACAAAGGTGCAATCATTGGACTCATGGTGGGCGGTGTTGTCA TAGCGACAGTGATCGTCATCACCTTGGTGATGCTGAAGAAGAAAC AGTACACATCCATTCATCATGGTGTGGTGGAGGTTGACGCCGCTGT CACCCCAGAGGAGCGCCACCTGTCCAAGATGCAGCAGAACGGCTA CGAAAATCCAACCTACAAGTTCTTTGAGCAGATGCAGAACTAG 11 ncAPP- ATGCTGCCCGGTTTGGCACTGCTCCTGCTGGCCGCCTGGACGGCTC R2/17 GGGCGCTGGAGGTACC C:A ATCATTGGACTCATGGTGGGCGGTGTT GTCATAGCGACAGTGATCGTCATCACCTTGGTGATGCTGAAGAAG AAACAGTACACATCCATTCATCATGGTGTGGTGGAGGTTGACGCC GCTGTCACCCCAGAGGAGCGCCACCTGTCCAAGATGCAGCAGAAC GGCTACGAAAATCCAACCTACAAGTTCTTTGAGCAGATGCAGAAC TAG 12 cAPP- ATGCTGCCCGGTTTGGCACTGCTCCTGCTGGCCGCCTGGACGGCTC R2/16 GGGCGCTGGAGGTACCCACTGATGGTAATGCTGGCCTGCTGGCTG AACCCCAGATTGCCATGTTCTGTGGCAGACTGAACATGCACATGA ATGTCCAGAATGGGAAGTGGGATTCAGATCCATCAGGGACCAAAA CCTGCATTGATACCAAGGAAGGCATCCTGCAGTA T:A TGCAGAATT CCGACATGACTCAGGATATGAAGTTCATCATCAAAAATTGGTGTTC TTTGCAGAAGATGTGGGTTCAAACAAAGGTGCAATCATTGGACTC ATGGTGGGCGGTGTTGTCATAGCGACAGTGATCGTCATCACCTTGG TGATGCTGAAGAAGAAACAGTACACATCCATTCATCATGGTGTGG TGGAGGTTGACGCCGCTGTCACCCCAGAGGAGCGCCACCTGTCCA AGATGCAGCAGAACGGCTACGAAAATCCAACCTACAAGTTCTTTG AGCAGATGCAGAACTAG 13 cAPP- ATGCTGCCCGGTTTGGCACTGCTCCTGCTGGCCGCCTGGACGGCTC R6/17 GGGCGCTGGAGGTACCCACTGATGGTAATGCTGGCCTGCTGGCTG AACCCCAGATTGCCATGTTCTGTGGCAGACTGAACATGCACATGA ATGTCCAGAATGGGAAGTGGGATTCAGATCCATCAGGGACCAAAA CCTGCATTGATACCAAGGAAGGCATCCTGCAGTATTGCCAAGAAG TCTACCCTGAACTGCAGATCACCAATGTGGTAGAAGCCAACCAAC CAGTGACCATCCAGAACTGGTGCAAGCGGGGCCGCAAGCAGTGCA AGACCCATCCCCACTTTGTGATTCCCTACCGCTGCTTAGTTGGTGA GTTTGTAAGTGATGCCCTTCTCGTTCCTGACAAGTGCAAATTCTTA CACCAGGAGAGGATGGATGTTTGCGAAACTCATCTTCACTGGCAC ACCGTCGCCAAAGAGACATGCAGTGAGAAGAGTACCAACTTGCAT GACTACGGCATGTTGCTGCCCTGCGGAATTGACAAGTTCCGAGGG GTAGAGTTTGTGTGTTGCCCACTGGCTGAAGAAAGTGACAATGTG GATTCTGCTGATGCGGAGGAGGATGACTCGGATGTCTGGTGGGGC GGAGCAGACACAGACTATGCAGATGGGAGTGAAGACAA A:G GTGC AATCATTGGACTCATGGTGGGCGGTGTTGTCATAGCGACAGTGATC GTCATCACCTTGGTGATGCTGAAGAAGAAACAGTACACATCCATT CATCATGGTGTGGTGGAGGTTGACGCCGCTGTCACCCCAGAGGAG CGCCACCTGTCCAAGATGCAGCAGAACGGCTACGAAAATCCAACC TACAAGTTCTTTGAGCAGATGCAGAACTAG 14 ncAPP- ATGCTGCCCGGTTTGGCACTGCTCCTGCTGGCCGCCTGGACGGCTC R2/14 GGGCGCTGGAGGTACCCACTGATGGTAATGCTGGCCTGCTGGCTG AACCCCAGATTGCCATGTTCTGTGGCAGACTGAACATGCACATGA ATGTCCAGAATGGGAAGTGGGATTCAGATCCATCAGGGACCAAAA CCTGCATTGATACCAA G:G ATCAGTTACGGAAACGATGCTCTCATG CCATCTTTGACCGAAACGAAAACCACCGTGGAGCTCCTTCCCGTGA ATGGAGAGTTCAGCCTGGACGATCTCCAGCCGTGGCATTCTTTTGG GGCTGACTCTGTGCCAGCCAACACAGAAAACGAAGTTGAGCCTGT TGATGCCCGCCCTGCTGCCGACCGAGGACTGACCACTCGACCAGG TTCTGGGTTGACAAATATCAAGACGGAGGAGATCTCTGAAGTGAA GATGGATGCAGAATTCCGACATGACTCAGGATATGAAGTTCATCA TCAAAAATTGGTGTTCTTTGCAGAAGATGTGGGTTCAAACAAAGG TGCAATCATTGGACTCATGGTGGGCGGTGTTGTCATAGCGACAGTG ATCGTCATCACCTTGGTGATGCTGAAGAAGAAACAGTACACATCC ATTCATCATGGTGTGGTGGAGGTTGACGCCGCTGTCACCCCAGAG GAGCGCCACCTGTCCAAGATGCAGCAGAACGGCTACGAAAATCCA ACCTACAAGTTCTTTGAGCAGATGCAGAACTAG 15 ncAPP- ATGCTGCCCGGTTTGGCACTGCTCCTGCTGGCCGCCTGGACGGCTC R14/17- GGGCGCTGGAGGTACCCACTGATGGTAATGCTGGCCTGCTGGCTG d8 AACCCCAGATTGCCATGTTCTGTGGCAGACTGAACATGCACATGA ATGTCCAGAATGGGAAGTGGGATTCAGATCCATCAGGGACCAAAA CCTGCATTGATACCAAGGAAGGCATCCTGCAGTATTGCCAAGAAG TCTACCCTGAACTGCAGATCACCAATGTGGTAGAAGCCAACCAAC CAGTGACCATCCAGAACTGGTGCAAGCGGGGCCGCAAGCAGTGCA AGACCCATCCCCACTTTGTGATTCCCTACCGCTGCTTAGTTGGTGA GTTTGTAAGTGATGCCCTTCTCGTTCCTGACAAGTGCAAATTCTTA CACCAGGAGAGGATGGATGTTTGCGAAACTCATCTTCACTGGCAC ACCGTCGCCAAAGAGACATGCAGTGAGAAGAGTACCAACTTGCAT GACTACGGCATGTTGCTGCCCTGCGGAATTGACAAGTTCCGAGGG GTAGAGTTTGTGTGTTGCCCACTGGCTGAAGAAAGTGACAATGTG GATTCTGCTGATGCGGAGGAGGATGACTCGGATGTCTGGTGGGGC GGAGCAGACACAGACTATGCAGATGGGAGTGAAGACAAAGTAGT AGAAGTAGCAGAGGAGGAAGAAGTGGCTGAGGTGGAAGAAGAAG AAGCCGATGATGACGAGGACGATGAGGATGGTGATGAGGTAGAG GAAGAGGCTGAGGAACCCTACGAAGAAGCCACAGAGAGAACCAC CAGCATTGCCACCACCACCACCACCACCACAGAGTCTGTGGAAGA GGTGGTTCGAGAGGTGTGCTCTGAACAAGCCGAGACGGGGCCGTG CCGAGCAATGATCTCCCGCTGGTACTTTGATGTGACTGAAGGGAA GTGTGCCCCATTCTTTTACGGCGGATGTGGCGGCAACCGGAACAA CTTTGACACAGAAGAGTACTGCATGGCCGTGTGTGGCAGCGCCAT TCCTACAACAGCAGCCAGTACCCCTGATGCCGTTGACAAGTATCTC GAGACACCTGGGGATGAGAATGAACATGCCCATTTCCAGAAAGCC AAAGAGAGGCTTGAGGCCAAGCACCGAGAGAGAATGTCCCAGGT CATGAGAGAATGGGAAGAGGCAGAACGTCAAGCAAAGAACTTGC CTAAAGCTGATAAGAAGGCAGTTATCCAGCATTTCCAGGAGAAAG TGGAATCTTTGGAACAGGAAGCAGCCAACGAGAGACAGCAGCTGG TGGAGACACACATGGCCAGAGTGGAAGCCATGCTCAATGACCGCC GCCGCCTGGCCCTGGAGAACTACATCACCGCTCTGCAGGCTGTTCC TCCTCGGCCTCGTCACGTGTTCAATATGCTAAAGAAGTATGTACGC GCAGAACAGAAGGACAGACAGCACACCCTAAAGCATTTCGAGCAT GTGCGCATGGTGGATCCCAAGAAAGCCGCTCAGATCCGGTCCCAG GTTATGACACTCCTCCGTGTGATTTATGAGCGCATGAATCAGTCTC TCTCCCTGCTCTACAACGTGCCTGCAGTGGCCGAGGAGATTCAGGA TGAAGTTGG T:G TTCTTTGCAGAAGATGTGGGTTCAAACAAAGGTG CAATCATTGGACTCATGGTGGGCGGTGTTGTCATAGCGACAGTGAT CGTCATCACCTTGGTGATGCTGAAGAAGAAACAGTACACATCCAT TCATCATGGTGTGGTGGAGGTTGACGCCGCTGTCACCCCAGAGGA GCGCCACCTGTCCAAGATGCAGCAGAACGGCTACGAAAATCCAAC CTACAAGTTCTTTGAGCAGATGCAGAACTAG 16 cAPP- ATGCTGCCCGGTTTGGCACTGCTCCTGCTGGCCGCCTGGACGGCTC D2/18-3 GGGCGCTGGAGGTACCCACTGATGGTAATGCTGGCCTGCTGGCTG AACCCCAGATTGCCATGTTCTGTGGCAG A:A AGTTCTTTGAGCAGA TGCAGAAC *Nomenclature: c, coding; nc, non-coding; R, RNA; D, DNA; X/Y, junction between exon X/Y; -X, number of variants. **X:Y indicate intraexonic junctions - In some embodiments, the non-classical variant of APP is a RNA sequence encoding for amyloid beta precursor protein or a portion thereof. In some embodiments, the non-classical variant of APP is a RNA sequence comprising intraexonic junctions between exons of APP. In some embodiments, the non-classical variant of APP is a RNA sequence lacking introns between exons of APP. In some embodiments, the non-classical variant of APP is a RNA sequence comprising one or more single nucleotide variant(s) of APP. In some embodiments, the RNA sequence is set forth as in Table 1. In some embodiments, the non-classical variant of APP is a DNA sequence encoding for the amyloid beta precursor protein or a portion thereof. In some embodiments, the non-classical variant of APP is a DNA sequence comprising intraexonic junctions between exons of APP or conventional spliced exon-exon junctions present in APP RNA. In some embodiments, the non-classical variant of APP is a DNA sequence lacking introns between exons of APP. In some embodiments, the non-classical variant of APP is a DNA sequence comprising one or more single nucleotide variant(s) of APP. In some embodiments, the non-classical variant of APP is a peptide sequence of a protein encoded by a whole or a part of the non-classical variant of APP.
- Provided herein, in certain embodiments, are non-classical variants of APP, wherein the non-classical variants comprise a single nucleotide variation (SNV). In some embodiments, the non-classical variants comprise one or more SNVs. In some embodiments, the SNV are familial mutations that arise somatically. In some embodiments, the SNV are associated with pathogenic Alzheimer's disease. Exemplary familial mutations associated with pathogenic Alzheimer's disease which translate to amino acid positions in APP include, but are not limited to, K670, M671, A673, D678, E682, K687, A692, E693, D694, A713, T714, V715, I716, V717, T719, M722, L723, and K724, wherein the amino acids correspond to
positions 670, 671, 673, 678, 682, 687, 692, 693, 694, 713, 714, 715, 716, 717, 719, 722, 723, and 724 of SEQ ID NO: 17. In some embodiments, the SNV in APP, which translate to amino acid positions in APP include, but are not limited to, K670N, M671L, A673V, D678H, D678N, E682K, K687N, A692G, E693G, D694N, A713T, T714A, T714I, V715A, V715M, I716F, I716M, I716T, I716V, V717F, V717G, V717I, V717L, T719P, M722K, L723P, and K724N. - In some embodiments, the non-classical variants comprise a SNV in at least one exon of APP. In some embodiments, the non-classical variants comprise one or more SNVs in at least one exon of APP. In some embodiments, the non-classical variants comprise the SNV in one or more exons of APP. In some embodiments, the non-classical variants comprise the SNV in
exon exon 17. In some embodiments, the non-classical variants comprise the SNV in the amyloid beta region of APP. In some embodiments, the SNV in APP, which translates to amino acid positions in APP includes, but are not limited to, K670, M671, A673, D678, E682, K687, A692, E693, D694, A713, T714, V715, I716, V717, T719, M722, L723, and K724, wherein the amino acids correspond topositions 670, 671, 673, 678, 682, 687, 692, 693, 694, 713, 714, 715, 716, 717, 719, 722, 723, and 724 of SEQ ID NO: 17. In some embodiments, the SNV in APP, which translates to amino acid positions in APP includes, but are not limited to, A673, A713, T714, V715, I716, V717, T719, and L723, wherein the amino acids correspond topositions 673, 713, 714, 715, 716, 717, 719, and 723 of SEQ ID NO: 17. In some embodiments, the SNV in APP, which translates to amino acid positions in APP includes, but are not limited to, K670N, M671L, A673V, D678H, D678N, E682K, K687N, A692G, E693G, D694N, A713T, T714A, T714I, V715A, V715M, I716F, I716M, I716T, I716V, V717F, V717G, V717I, V717L, T719P, M722K, L723P, and K724N. In some embodiments, the SNV in APP, which translates to amino acid positions in APP includes, but are not limited to, A673V, A713T, T714I, V715M, V715A, I716M, V717I, V717F, T719P, and L723P. -
TABLE 2 Amino Acid Sequence of APP SEQ ID Accession NO No. Amino Acid Sequence 17 P05067.3 MLPGLALLLLAAWTARALEVPTDGNAGLLAEPQ IAMFCGRLNMHMNVQNGKWDSDPSGTKTCIDTK EGILQYCQEVYPELQITNVVEANQPVTIQNWCK RGRKQCKTHPHFVIPYRCLVGEFVSDALLVPDK CKFLHQERMDVCETHLHWHTVAKETCSEKSTNL HDYGMLLPCGIDKFRGVEFVCCPLAEESDNVDS ADAEEDDSDVWWGGADTDYADGSEDKVVEVAEE EEVAEVEEEEADDDEDDEDGDEVEEEAEEPYEE ATERTTSIATTTTTTTESVEEVVREVCSEQAET GPCRAMISRWYFDVTEGKCAPFFYGGCGGNRNN FDTEEYCMAVCGSAMSQSLLKTTQEPLARDPVK LPTTAASTPDAVDKYLETPGDENEHAHFQKAKE RLEAKHRERMSQVMREWEEAERQAKNLPKADKK AVIQHFQEKVESLEQEAANERQQLVETHMARVE AMLNDRRRLALENYITALQAVPPRPRHVFNMLK KYVRAEQKDRQHTLKHFEHVRMVDPKKAAQIRS QVMTHLRVIYERMNQSLSLLYNVPAVAEEIQDE VDELLQKEQNYSDDVLANMISEPRISYGNDALM PSLTETKTTVELLPVNGEFSLDDLQPWHSFGAD SVPANTENEVEPVDARPAADRGLTTRPGSGLTN IKTEEISEVKMDAEFRHDSGYEVHHQKLVFFAE DVGSNKGAIIGLMVGGVVIATVIVITLVMLKKK QYTSIHHGVVEVDAAVTPEERHLSKMQQNGYEN PTYKFFEQMQN - In some embodiments, the non-classical variant of APP comprises a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16. In some embodiments, the non-classical variant of APP comprises at least or about 70%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID NO: 1. In some embodiments, the non-classical variant of APP comprises at least or about 70%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID NO: 2. In some embodiments, the non-classical variant of APP comprises at least or about 70%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID NO: 3. In some embodiments, the non-classical variant of APP comprises at least or about 70%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID NO: 4. In some embodiments, the non-classical variant of APP comprises at least or about 70%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID NO: 5. In some embodiments, the non-classical variant of APP comprises at least or about 70%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID NO: 6. In some embodiments, the non-classical variant of APP comprises at least or about 70%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID NO: 7. In some embodiments, the non-classical variant of APP comprises at least or about 70%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID NO: 8. In some embodiments, the non-classical variant of APP comprises at least or about 70%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID NO: 9. In some embodiments, the non-classical variant of APP comprises at least or about 70%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID NO: 10. In some embodiments, the non-classical variant of APP comprises at least or about 70%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID NO: 11. In some embodiments, the non-classical variant of APP comprises at least or about 70%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID NO: 12. In some embodiments, the non-classical variant of APP comprises at least or about 70%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID NO: 13. In some embodiments, the non-classical variant of APP comprises at least or about 70%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID NO: 14. In some embodiments, the non-classical variant of APP comprises at least or about 70%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID NO: 15. In some embodiments, the non-classical variant of APP comprises at least or about 70%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID NO: 16.
- Methods of Identifying Non-classical Variants
- Described herein are methods for identifying non-classical variants of amyloid precursor protein (APP) gene. In some embodiments, a non-classical variant of APP is identified from an individual with Alzheimer's disease (AD). In some embodiments, a non-classical variant of APP is identified from a brain of an individual with AD. In some embodiments, a non-classical variant of APP is identified in a neuron isolated from an individual with AD. In some embodiments, the neuron is sorted and isolated. In some embodiments, the neuron is sorted and isolated using fluorescence activated nuclear sorting (FANS). In some embodiments, a non-classical variant of APP is identified from RNA extracted from a neuron of an individual with AD. In some embodiments, the RNA is reverse transcribed to cDNA, and a non-classical variant of APP is identified. In some embodiments, a non-classical variant of APP is identified from genomic DNA extracted from a neuron of an individual with AD. In some embodiments, AD is sporadic AD. In some embodiments, AD is familial AD. In some embodiments, a non-classical variant is identified using a control. In some embodiments, the control is an individual that does not have AD.
- In some embodiments, a non-classical variant of APP is identified using probes that hybridize to full length APP. In some embodiments, a non-classical variant of APP is identified using probes that hybridize to full length APP comprising only exons of APP. In some embodiments, the probes hybridize to genomic DNA. In some embodiments, the probes are used for in situ hybridization. In some embodiments, the probes hybridize to cDNA of APP. An exemplary probe comprises a sequence as set forth in SEQ ID NO: 21. In some embodiments, the probes hybridize to cDNA of APP and are detected by Southern blot. In some embodiments, the Southern Blot is compared to a gel electrophoresis of cDNA of APP. In some embodiments, a non-classical variant of APP is identified by sequencing cDNA corresponding to cDNA detected by the Southern Blot.
- Described herein, in certain embodiments, are methods for detecting one or more non-classical variant(s) of amyloid precursor protein (APP) gene in an individual in need thereof. Further described herein, in certain embodiments, are methods for evaluating an individual for risk of developing a disease or disorder characterized by unwanted accumulation of amyloid beta protein comprising: measuring an expression profile or an activity profile of one or more non-classical variant(s) of APP gene from a biological sample from the individual; comparing the expression profile or the activity profile of the one or more non-classical variant(s) of APP to a reference expression profile or activity profile of the one or more non-classical variant(s) of APP derived from a cohort of control individuals; and identifying the individual as having or not having a risk factor for developing the disease or disorder characterized by unwanted accumulation of amyloid beta protein based on a comparison of the expression profile or activity profile to the reference expression profile or activity profile of the cohort of control individuals. Further described herein, in certain embodiments, are methods of diagnosing a disease or disorder in an individual characterized by unwanted accumulation of amyloid beta protein, comprising: identifying the individual as having the disease or disorder characterized by unwanted accumulation of amyloid beta protein by comparing an expression profile or an activity profile of one or more non-classical variant(s) of an APP gene to a reference expression profile or activity profile of the one or more non-classical variant(s) of APP derived from a cohort of control individuals. In some embodiments, the biological sample is a blood sample. In some embodiments, the biological sample comprises RNA, DNA, or protein. In some embodiments, the disease or disorder is Alzheimer's disease. In some embodiments, the Alzheimer's disease is familial Alzheimer's disease (FAD) or sporadic Alzheimer's disease.
- In some embodiments, the amyloid beta protein is an amyloidogenic protein or a protein that produces an amyloid like morphology. In some embodiments, the amyloid beta protein is encoded by the one or more non-classical variant(s) of APP. In some embodiments, the amyloid beta protein is a variant peptide or protein comprising amyloid beta that is encoded by the one or more non-classical variant(s) of APP. In some embodiments, the amyloid beta protein is a variant peptide or protein that does not comprise amyloid beta and is encoded by the one or more non-classical variant(s) of APP.
- In some embodiments, the expression profile is expression level of the one or more non-classical variant(s) of APP. In some embodiments, the expression profile is expression of a set of different non-classical variant of APP. In some embodiments, the activity profile is activity level of the one or more non-classical variant(s) of APP. In some embodiments, the activity profile is activity of a set of different non-classical variant of APP. In some embodiments, the non-classical variant of APP comprises a portion or all of an exon of the APP gene. In some embodiments, the non-classical variant of APP comprises a portion or all of
exon exon comprise exon 8. In some embodiments, the one or more exon(s) in the one or more non-classical variant(s) of APP is rearranged as compared to control. In some embodiments, the non-classical variant of APP does not compriseexon 8 of the APP gene. In some embodiments, the non-classical variant of APP comprises a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16. In some embodiments, the non-classical variant of APP comprises a single nucleotide variation (SNV). In some embodiments, the non-classical variant of APP comprises one or more SNVs. In some embodiments, the SNV in APP, which translates to amino acid positions in APP includes, but are not limited to, K670, M671, A673, D678, E682, K687, A692, E693, D694, A713, T714, V715, I716, V717, T719, M722, L723, and K724, wherein the amino acids correspond topositions 670, 671, 673, 678, 682, 687, 692, 693, 694, 713, 714, 715, 716, 717, 719, 722, 723, and 724 of SEQ ID NO: 17. In some embodiments, the SNV in APP, which translates to amino acid positions in APP includes, but are not limited to, A673, A713, T714, V715, I716, V717, T719, and L723, wherein the amino acids correspond topositions 673, 713, 714, 715, 716, 717, 719, and 723 of SEQ ID NO: 17. In some embodiments, the SNV in APP, which translates to amino acid positions in APP includes, but are not limited to, K670N, M671L, A673V, D678H, D678N, E682K, K687N, A692G, E693G, D694N, A713T, T714A, T714I, V715A, V715M, I716F, I716M, I716T, I716V, V717F, V717G, V717I, V717L, T719P, M722K, L723P, and K724N. In some embodiments, the SNV in APP, which translates to amino acid positions in APP includes, but are not limited to, A673V, A713T, T714I, V715M, V715A, I716M, V717I, V717F, T719P, and L723P. - Sequencing
- In some embodiments, the expression profile of one or more non-classical variant(s) of amyloid precursor protein (APP) gene is measured by sequencing of a biological sample from the individual. In some embodiments, the biological sample comprises RNA or DNA. In some embodiments, sequencing is performed with any appropriate sequencing technology, including but not limited to single molecule real-time sequencing, Polony sequencing, sequencing by ligation, reversible terminator sequencing, proton detection sequencing, ion semiconductor sequencing, nanopore sequencing, electronic sequencing, pyrosequencing, Maxam-Gilbert sequencing, chain termination (e.g., Sanger) sequencing, +S sequencing, or sequencing by synthesis.
- In some embodiments, the expression profile of one or more non-classical variant(s) of APP is measured by nucleotide sequencing. In some embodiments, the nucleotide sequencing comprises long-read sequencing. In some embodiments, the long-read sequencing is single molecule real-time sequencing. In some embodiments, the single molecule real-time sequencing comprises attaching a different fluorescent dye to each of the nucleic acid bases and using a polymerase. In some embodiments, the polymerase incorporates a single nucleotide comprising the fluorescent dye. In some embodiments, the fluorescent dye is detected to generate a nucleotide sequence. In some embodiments, the polymerase is a DNA polymerase or a RNA polymerase. In some embodiments, the DNA polymerase is a modified polymerase. In some embodiments, a template to be sequenced is a DNA template or a RNA template. In some embodiments, the long-read sequencing does not require a reference genome. In some embodiments, methods for measuring the expression profile comprise preparation of a biological sample prior to sequencing. In some embodiments, DNA is extracted and purified from the biological sample. In some embodiments, RNA is extracted. In some embodiments, RNA is extracted, purified, and reverse transcribed to cDNA. In some embodiments, after RNA or DNA is extracted, the reverse transcribed cDNA or DNA is amplified prior to sequencing. In some embodiments, single molecule real-time sequencing comprises additional preparation of the biological sample prior to sequencing. In some embodiments, the DNA is fragmented. In some embodiments, target regions are amplified to obtain fragmented DNA. In some embodiments, target regions are enriched by hybridization based DNA pull-down. Following fragmentation, in some embodiments, the ends of the DNA are repaired. In some embodiments, hairpin adapters are ligated to the DNA that then hybridizes to a primer. In some embodiments, a nuclease is used to remove DNA that did not ligate to the hairpin adapters. In some embodiments, a DNA polymerase is mixed and the DNA is sequenced.
- In some embodiments, the expression profile of one or more non-classical variant(s) of APP is the expression level of one or more non-classical variant(s) of APP. In some embodiments, the expression level is of a set of different non-classical variants of APP. In some embodiments, the expression level is measured following long-read sequencing. In some embodiments, the long-read sequencing is RNA sequencing. In some embodiments, the long-read sequencing is DNA sequencing. In some embodiments, the long-read sequencing is single molecule real-time sequencing. During a long-read sequencing reaction, sequenced base pairs or “reads” are generated. In some embodiments, the expression level is then quantified by counting a number of reads that map to the one or more non-classical variant(s) of APP sequences during the long-read sequencing reaction. In some embodiments, the one or more non-classical variant(s) of APP sequences comprise a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16.
- In some embodiments, the long read sequencing measures a change in the APP gene. In some embodiments, the change is a change in DNA of the APP gene. In some embodiments, the DNA is coding DNA. In some embodiments, the DNA is non-coding DNA. In some embodiments, the change is a change in RNA of the APP gene. In some embodiments, the RNA is coding RNA. In some embodiments, the RNA is non-coding RNA. In some embodiments, the change is a change in a protein encoded by the APP gene.
- In some embodiments, the long read sequencing measures a single nucleotide variation (SNV) in the APP gene. In some embodiments, the long read sequencing measures one or more SNVs in the APP gene. In some embodiments, the SNV in APP, which translates to amino acid positions in APP includes, but are not limited to, K670, M671, A673, D678, E682, K687, A692, E693, D694, A713, T714, V715, I716, V717, T719, M722, L723, and K724, wherein the amino acids correspond to
positions 670, 671, 673, 678, 682, 687, 692, 693, 694, 713, 714, 715, 716, 717, 719, 722, 723, and 724 of SEQ ID NO: 17. In some embodiments, the SNV in APP, which translates to amino acid positions in APP includes, but are not limited to, A673, A713, T714, V715, I716, V717, T719, and L723, wherein the amino acids correspond topositions 673, 713, 714, 715, 716, 717, 719, and 723 of SEQ ID NO: 17. In some embodiments, the SNV in APP, which translates to amino acid positions in APP includes, but are not limited to, K670N, M671L, A673V, D678H, D678N, E682K, K687N, A692G, E693G, D694N, A713T, T714A, T714I, V715A, V715M, I716F, I716M, I716T, I716V, V717F, V717G, V717I, V717L, T719P, M722K, L723P, and K724N. In some embodiments, the SNV in APP, which translates to amino acid positions in APP includes, but are not limited to, A673V, A713T, T714I, V715M, V715A, I716M, V717I, V717F, T719P, and L723P. - Pull-Down Assays
- In some embodiments, the expression profile of one or more non-classical variant(s) of the amyloid precursor protein (APP) gene is measured by a pull-down assay. In some embodiments, one or more probe(s) for use in the pull-down assay is designed to hybridize to a portion or all of
exon exon - In some embodiments, the one or more probe(s) is labeled with an affinity tag. Exemplary affinity tags include, but are not limited to, biotin, desthiobiotin, histidine, polyhistidine, myc, hemagglutinin (HA), FLAG, glutathione S transferase (GST), or derivatives thereof. In some embodiments, the affinity tag is recognized by avidin, streptavidin, nickel, or glutathione.
- In some embodiments, following a pull-down assay, one or more non-classical variant(s) of APP are amplified. In some embodiments, the one or more non-classical variant(s) of APP are amplified using primers designed to detect
exon exon - Following the pull-down assay, in some embodiments, the one or more non-classical variant(s) of APP are not amplified. In some embodiments, the one or more non-classical variant(s) of APP are visualized using a fluorescent assay, a radioactivity assay, or a luminescent assay. For example, the one or more probes used to hybridize to the one or more non-classical variant(s) of APP further comprises a fluorescent tag that is detected using the fluorescent assay. In some embodiments, the one or more non-classical variant(s) of APP are visualized by gel electrophoresis.
- In some embodiments, the pull down assay measures a change in the APP gene. In some embodiments, the change is a change in DNA of the APP gene. In some embodiments, the DNA is coding DNA. In some embodiments, the DNA is non-coding DNA. In some embodiments, the change is a change in RNA of the APP gene. In some embodiments, the RNA is coding RNA. In some embodiments, the RNA is non-coding RNA. In some embodiments, the change is a change in a protein encoded by the APP gene.
- Hybridization Assays
- In some embodiments, the expression profile of one or more non-classical variant(s) of the amyloid precursor protein (APP) gene is measured by binding of one or probe(s) to one or more non-classical variant(s) of APP. In some embodiments, the one or more probe(s) is a polypeptide. In some embodiments, the one or more probe(s) is a polynucleotide.
- In some embodiments, intraexonic rearrangements are detected by measuring binding of the one or more probe(s). In some embodiments, the one or more probe(s) hybridizes to target sequences within at least two exons of APP gene. For example, the one or more probe(s) hybridizes to sequences with one end complementary to a 3′ end of one exon of APP and a second end complementary to a 5′ end of a downstream exon of APP. In some embodiments, the one or more probe(s) hybridizes to target sequences within at least two exons that are consecutive exons of APP. In some embodiments, the one or more probe(s) hybridizes to target sequences within at least two exons that are non-consecutive exons of APP.
- In some embodiments, the one or more probe(s) are provided in an array. In some embodiments, the array comprises one or more probe(s) for measuring an expression profile for one or more non-classical variant(s) of APP. In some embodiments, the one or more probe(s) detects RNA. In some embodiments, the one or more probe(s) detect exonic rearrangements such as intraexonic rearrangements of APP. In some embodiments, the one or more probes hybridize to a portion or all of an
exon exon - In some embodiments, the one or more probe(s) bind to RNA or DNA from the one or more non-classical variant(s) of APP gene. In some embodiments, the one or more probe(s) are used for an amplification reaction. In some embodiments, the amplification reaction is PCR. In some embodiments, the amplification reaction is quantitative such as qPCR. In some embodiments, the PCR reaction utilizes a TaqMan™ or a similar quantitative PCR technology. In some embodiments, at least one primer used in the PCR reaction comprises a sequence as set forth in SEQ ID NO: 19 or 20.
- The one or more probe(s), in some embodiments, bind to a protein encoded by the one or more non-classical variant(s) of APP gene. Exemplary methods for detecting binding of the one or more probe(s) include, but are not limited to, enzyme linked immunosorbent assays (ELISA), Western blots, spectroscopy, mass spectrometry, peptide arrays, colorimetry, electrophoresis, isoelectric focusing, immunoprecipitations, immunoassays, and immunofluorescence.
- In some embodiments, the expression profile of the one or more non-classical variant(s) of the APP gene is measured by in situ hybridization or immunological hybridization. In some embodiments, the in situ hybridization is performed without amplification. In some embodiments, the in situ hybridization is performed without polymerase dependent amplification. In some embodiments, the in situ hybridization comprises capturing the one or more non-classical variant(s) of APP from the biological sample on a solid support prior to contacting the one or more non-classical variant(s) of APP with the one or more probe(s). In some embodiments, the in situ hybridization is chromogenic in situ hybridization. In some embodiments, the in situ hybridization is fluorescence in situ hybridization. In some embodiments, in situ hybridization allows for detection of intraexonic rearrangements. In some embodiments, in situ hybridization allows for detection for genomic rearrangements such as between introns and exons or between exons and exons. In some embodiments, the one or more probe(s) for in situ hybridization hybridizes to a region spanning an intraexonic rearrangement. For example, for the non-classical variant cAPP-R3/16, the one or more probe(s) hybridize to a
region spanning exon 3 andexon 16. In some embodiments, the one or more probe(s) hybridize to a region of a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16. - Described herein, in certain embodiments, one or more probe(s) that hybridize to an intraexonic junction are non-classical variants of APP comprising portions of at least two exons, wherein the at least two exons are linked by intraexonic junctions. In some embodiments, the one or more probe(s) hybridize to an intraexonic junction between
exon 1 andexon exon 2 andexon exon 3 andexon exon 4 andexon exon 5 andexon exon 6 andexon exon 7 andexon exon 8 andexon exon 9 andexon exon 10 andexon exon 11 andexon exon 12 andexon exon 13 andexon exon 14 andexon exon 15 andexon exon 16 andexon exon 17 andexon exon 18 andexon exon 1 andexon 11,exon 1 andexon 14,exon 2 andexon 17,exon 2 andexon 14,exon 2 andexon 18,exon 2 andexon 16,exon 3 andexon 16,exon 3 andexon 14,exon 3 andexon 17,exon 6 andexon 17,exon 6 andexon 18,exon 3 andexon 9,exon 2 andexon 9,exon 16 andexon 18,exon 6 andexon 12,exon 5 andexon 16, orexon 16 andexon 17. - In situ hybridization, in some embodiments, comprises probes for detecting one or more non-classical variant(s) of APP. In some embodiments, the probes hybridize to RNA comprising the one or more non-classical variant(s) of APP. In some embodiments, the probes hybridize to DNA comprising the one or more non-classical variant(s) of APP. In some embodiments, the probes hybridize to RNA comprising a portion of a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16. In some embodiments, the probes comprise a base-pairing region complementary to the target nucleic acid, a spacer sequence, and a base-tail sequence. In some embodiments, two tail sequences form a hybridization site for amplification. In some embodiments, the target nucleic acid is visualized following amplification. In some embodiments, the probes hybridize to protein encoded by the non-classical variant of APP. In some embodiments, the probes are removed prior to visualization. In some embodiments, the probes are removed enzymatically, chemically, or mechanically. For example, the probes are removed using restriction enzymes.
- In some embodiments, a number of probes are used for in situ hybridization. In some embodiments, in situ hybridization comprises at least or about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100 or more 100 probes. In some embodiments, in situ hybridization comprises at most 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, or 100 probes. In some embodiments, a number of probe pairs are used for in situ hybridization. In some embodiments, the number of probe pairs comprises at least or about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 45, 50, or more than 50 probe pairs. In some embodiments, in situ hybridization comprises at most 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 45, or 50 probe pairs. In some embodiments, following hybridization of a probe or a probe pair, the probe or probe pair are amplified prior to visualization.
- In some embodiments, a probe is labeled. In some embodiments, a probe is labeled with a radioactive label, a fluorescent label, an enzyme, a chemiluminescent tag, a colorimetric tag, an affinity tag or other labels or tags that are known in the art.
- Exemplary affinity tags include, but are not limited to, biotin, desthiobiotin, histidine, polyhistidine, myc, hemagglutinin (HA), FLAG, glutathione S transferase (GST), or derivatives thereof. In some embodiments, the affinity tag is recognized by avidin, streptavidin, nickel, or glutathione.
- In some embodiments, the fluorescent label is a fluorophore, a fluorescent protein, a fluorescent peptide, quantum dots, a fluorescent dye, a fluorescent material, or variations or combinations thereof.
- Exemplary fluorophores include, but are not limited to, Alexa-Fluor dyes (e.g., Alexa Fluor® 350, Alexa Fluor® 405, Alexa Fluor® 430, Alexa Fluor® 488,
Alexa Fluor® 500, Alexa Fluor® 514, Alexa Fluor® 532, Alexa Fluor® 546, Alexa Fluor® 555, Alexa Fluor® 568, Alexa Fluor® 594, Alexa Fluor® 610, Alexa Fluor® 633, Alexa Fluor® 647, Alexa Fluor® 660, Alexa Fluor® 680, Alexa Fluor® 700, and Alexa Fluor® 750), APC, Cascade Blue, Cascade Yellow and R-phycoerythrin (PE), DyLight 405, DyLight 488, DyLight 550, DyLight 650, DyLight 680, DyLight 755, DyLight 800, FITC, Pacific Blue, PerCP, Rhodamine, and Texas Red, Cy5, Cy5.5, Cy7. - Examples of fluorescent peptides include GFP (Green Fluorescent Protein) or derivatives of GFP (e.g., EBFP, EBFP2, Azurite, mKalamal, ECFP, Cerulean, CyPet, YFP, Citrine, Venus, YPet).
- Examples of fluorescent dyes include, but are not limited to, xanthenes (e.g., rhodamines, rhodols and fluoresceins, and their derivatives); bimanes; coumarins and their derivatives (e.g., umbelliferone and aminomethyl coumarins); aromatic amines (e.g., dansyl; squarate dyes); benzofurans; fluorescent cyanines; indocarbocyanines; carbazoles; dicyanomethylene pyranes; polymethine; oxabenzanthrane; xanthene; pyrylium; carbostyl; perylene; acridone; quinacridone; rubrene; anthracene; coronene; phenanthrecene; pyrene; butadiene; stilbene; porphyrin; pthalocyanine; lanthanide metal chelate complexes; rare-earth metal chelate complexes; and derivatives of such dyes. In some embodiments, the fluorescein dye is, but not limited to, 5-carboxyfluorescein, fluorescein-5-isothiocyanate, fluorescein-6-isothiocyanate and 6-carboxyfluorescein. In some embodiments, the rhodamine dye is, but not limited to, tetramethylrhodamine-6-isothiocyanate, 5-carboxytetramethylrhodamine, 5-carboxy rhodol derivatives, tetramethyl and tetraethyl rhodamine, diphenyldimethyl and diphenyldiethyl rhodamine, dinaphthyl rhodamine, and rhodamine 101 sulfonyl chloride (sold under the tradename of TEXAS RED®). In some embodiments, the cyanine dye is Cy3, Cy3B, Cy3.5, Cy5, Cy5.5, Cy7, IRDYE680, Alexa Fluor 750, IRDye800CW, or ICG.
- Fluorescent labels are detected by any suitable method. For example, a fluorescent label is detected by exciting the fluorochrome with the appropriate wavelength of light and detecting the resulting fluorescence, e.g., by microscopy, visual inspection, via photographic film, by the use of electronic detectors such as charge coupled devices (CCDs), or photomultipliers. In some embodiments, the one or more probe(s) are labeled with the same fluorescent label. In some embodiments, the one or more probe(s) are labeled with different fluorescent labels.
- In some embodiments, the expression profile is expression level of the one or more non-classical variant(s) of APP. In some embodiments, the expression level is of a set of different non-classical variants of APP. In some embodiments, the expression level is measured using PCR. Examples of PCR techniques include, but are not limited to quantitative PCR (qPCR), single cell PCR, PCR-RFLP, digital PCR (dPCR), droplet digital PCR (ddPCR), single marker qPCR, hot start PCR, and Nested PCR.
- In some embodiments, the expression level is measured using qPCR. In some embodiments, the qPCR comprises use of fluorescent dyes or fluorescent probes. In some embodiments, the fluorescent dye is an intercalating dye. Examples of intercalating dyes include, but are not limited to, intercalating dyes include SYBR green I, SYBR green II, SYBR gold, ethidium bromide, methylene blue, Pyronin Y, DAPI, acridine orange, Blue View, or phycoerythrin. In some embodiments, the qPCR comprises use of more than one fluorescent probe. In some embodiments, the use of more than one fluorescent probes allows for multiplexing. For example, different non-classical variants are hybridized to different fluorescent probes and can be detected in a single qPCR reaction.
- In some embodiments, the probe is used for visualization of the one or more non-classical variant(s) of APP in an individual. In some embodiments, the probe is visualized by X-Ray, fluoroscopes, ultrasound, CT-scan, PET scan, magnetic resonance image (MRIs), or electromagnetic field.
- In some embodiments, the hybridization assay measures a change in the APP gene. In some embodiments, the change is a change in DNA of the APP gene. In some embodiments, the DNA is coding DNA. In some embodiments, the DNA is non-coding DNA. In some embodiments, the change is a change in RNA of the APP gene. In some embodiments, the RNA is coding RNA. In some embodiments, the RNA is non-coding RNA. In some embodiments, the change is a change in a protein encoded by the APP gene.
- Samples
- Described herein, in certain embodiments are methods for diagnosing an individual having or suspected of having Alzheimer's disease by measuring the expression profile or the activity profile of the one or more non-classical variant(s) of amyloid precursor protein (APP) gene. In some embodiments, the expression profile or the activity profile of the one or more non-classical variant(s) of APP is associated with unwanted accumulation of amyloid beta protein but the individual does not have Alzheimer's disease.
- In some embodiments, the expression profile or the activity profile is determined from a biological sample from the individual. In some embodiments, the biological sample comprises RNA or DNA. In some embodiments, the RNA is pre-mRNA. In some embodiments, the RNA is mRNA. In some embodiments, the DNA is nuclear DNA. In some embodiments, the DNA is extrachromosomal or extranuclear DNA. In some embodiments, the DNA is circular DNA.
- In some embodiments, the biological sample is from a blood sample. The blood sample is taken, for example, from the individual by a blood draw. In some embodiments, the blood sample is processed by centrifugation such as by density centrifugation. In some embodiments, the blood sample is treated with a red blood cell lysis agent. In some embodiments, the blood sample comprises cells from the Central Nervous System (e.g., neurons, astrocytes, or microglia) that are released during break down of the blood brain barrier. In some embodiments, the biological sample is from cerebrospinal fluid. In some embodiments, the cerebrospinal fluid comprises cells from the Central Nervous System (e.g., neurons, astrocytes, or microglia) that are released during break down of the blood brain barrier.
- A biological sample, in some embodiments, comprises exosomes. Exosomes are cell-derived vesicles that are released from many cell types including, but not limited to, dendritic cells (DCs), lymphocytes, platelets, mast cells, epithelial cells, endothelial cells, and neurons. In some embodiments, the exosomes are found in blood. In some embodiments, the exosomes are found in cerebrospinal fluid. In some embodiments, the biological sample comprises exosomes from the blood. In some embodiments, the biological sample comprises exosomes from cerebrospinal fluid.
- In some embodiments, nucleic acid is extracted from the biological sample. In some embodiments, the nucleic acid is DNA. In some embodiments, the DNA is genomic DNA. In some embodiments, the DNA is extrachromosomal DNA. In some embodiments, the DNA is circular DNA. In some embodiments, the nucleic acid is RNA. The nucleic acid, in some embodiments, is extracted using any technique that does not interfere with subsequent analysis. For example, the nucleic acid is extracted using alcohol precipitation using ethanol, methanol, or isopropyl alcohol. In some embodiments, the nucleic acid is extracted using phenol, chloroform, or any combination thereof. In some embodiments, the nucleic acid is extracted using cesium chloride. In some embodiments, the nucleic acid is extracted using sodium, potassium or ammonium acetate or any other salt commonly used to precipitate DNA. In some embodiments, the nucleic acid is extracted using utilizes a column or resin based nucleic acid purification. In some embodiments, after extraction the nucleic acid is stored in water, Tris buffer, or Tris-EDTA buffer before subsequent analysis. For example, storage is less than 8° C., 4° C., −20° C., or −70° C. In some embodiments, the nucleic acid is stored for 1, 2, 3, 4, 5, 6, or 7 days. In some embodiments, the nucleic acid is stored for 1, 2, 3, or 4 weeks. In some embodiments, the nucleic acid is stored for 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 months.
- Described herein, in certain embodiments, are methods for evaluating an individual for risk of developing a disease or disorder characterized by unwanted accumulation of amyloid beta protein from a biological sample. In some embodiments, methods for evaluating the individual comprise measuring the expression profile or the activity profile of the one or more non-classical variant(s) of APP. In some embodiments, the one or more non-classical variant(s) of APP comprise a portion or all of an exon of APP gene. For example, the one or more non-classical variant(s) of APP comprise a portion or all of
exon exon exon 8. In some embodiments, the one or more non-classical variant(s) of APP comprise a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16. In some embodiments, the non-classical variants comprise a single nucleotide variation (SNV) in APP, which translate to amino acid positions in APP include, but are not limited to, K670, M671, A673, D678, E682, K687, A692, E693, D694, A713, T714, V715, I716, V717, T719, M722, L723, and K724, wherein the amino acids correspond topositions 670, 671, 673, 678, 682, 687, 692, 693, 694, 713, 714, 715, 716, 717, 719, 722, 723, and 724 of SEQ ID NO: 17. In some embodiments, the SNV in APP, which translate to amino acid positions in APP include, but are not limited to, A673, A713, T714, V715, I716, V717, T719, and L723, wherein the amino acids correspond topositions 673, 713, 714, 715, 716, 717, 719, and 723 of SEQ ID NO: 17. In some embodiments, the SNV in APP, which translate to amino acid positions in APP include, but are not limited to, K670N, M671L, A673V, D678H, D678N, E682K, K687N, A692G, E693G, D694N, A713T, T714A, T714I, V715A, V715M, I716F, I716M, I716T, I716V, V717F, V717G, V717I, V717L, T719P, M722K, L723P, and K724N. In some embodiments, the SNV in APP, which translate to amino acid positions in APP include, but are not limited to, A673V, A713T, T714I, V715M, V715A, I716M, V717I, V717F, T719P, and L723P. In some embodiments, the non-classical variants comprise one or more SNVs. In some embodiments, the expression profile is measured by long-read sequencing. In some embodiments, the long read-sequencing is single molecule real-time sequencing. In some embodiments, the expression profile is measured using probe-based assays. For example, the expression profile is measured by quantitative-PCR, in situ hybridization, or pull down assays. - Further described herein, in certain embodiments, are methods for detecting one or more non-classical variant(s) of an APP gene in an individual in need thereof, comprising: detecting an expression profile or an activity profile of the one or more non-classical variant(s) of the APP gene in a biological sample from the individual by a method comprising long-read sequencing of the biological sample. In some embodiments, the biological sample is a blood sample. In some embodiments, the biological sample comprises RNA or DNA. In some embodiments, the one or more non-classical variant(s) of APP comprise a portion or all of an exon of APP gene. In some embodiments, the one or more non-classical variant(s) of APP comprise a portion or all of
exon exon exon 8. In some embodiments, the non-classical variants comprise a single nucleotide variation (SNV) in APP, which translate to amino acid positions in APP include, but are not limited to, K670, M671, A673, D678, E682, K687, A692, E693, D694, A713, T714, V715, I716, V717, T719, M722, L723, and K724, wherein the amino acids correspond topositions 670, 671, 673, 678, 682, 687, 692, 693, 694, 713, 714, 715, 716, 717, 719, 722, 723, and 724 of SEQ ID NO: 17. In some embodiments, the SNV in APP, which translate to amino acid positions in APP include, but are not limited to, A673, A713, T714, V715, I716, V717, T719, and L723, wherein the amino acids correspond topositions 673, 713, 714, 715, 716, 717, 719, and 723 of SEQ ID NO: 17. In some embodiments, the SNV in APP, which translate to amino acid positions in APP include, but are not limited to, K670N, M671L, A673V, D678H, D678N, E682K, K687N, A692G, E693G, D694N, A713T, T714A, T714I, V715A, V715M, I716F, I716M, I716T, I716V, V717F, V717G, V717I, V717L, T719P, M722K, L723P, and K724N. In some embodiments, the SNV in APP, which translate to amino acid positions in APP include, but are not limited to, A673V, A713T, T714I, V715M, V715A, I716M, V717I, V717F, T719P, and L723P. In some embodiments, the one or more non-classical variant(s) of APP comprise a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16. - Further described herein, in certain embodiments, are methods of detecting one or more non-classical variant(s) of an APP gene in an individual in need thereof, comprising: detecting an expression profile or an activity profile of the one or more non-classical variant(s) of the APP gene in the biological sample from the individual by a method comprising binding of one or more probe(s) to the one or more non-classical variant(s) of APP. In some embodiments, the biological sample is a blood sample. In some embodiments, the biological sample comprises RNA, DNA, or protein. In some embodiments, the one or more non-classical variant(s) of APP comprise a portion or all of an exon of APP gene. In some embodiments, the one or more non-classical variant(s) of APP comprise a portion or all of
exon exon exon 8. In some embodiments, the single nucleotide variation (SNV) in APP, which translates to amino acid positions in APP includes, but are not limited to, K670, M671, A673, D678, E682, K687, A692, E693, D694, A713, T714, V715, I716, V717, T719, M722, L723, and K724, wherein the amino acids correspond topositions 670, 671, 673, 678, 682, 687, 692, 693, 694, 713, 714, 715, 716, 717, 719, 722, 723, and 724 of SEQ ID NO: 17. In some embodiments, the SNVs in APP, which translate to amino acid positions in APP include, but are not limited to, A673, A713, T714, V715, I716, V717, T719, and L723, wherein the amino acids correspond topositions 673, 713, 714, 715, 716, 717, 719, and 723 of SEQ ID NO: 17. In some embodiments, the SNVs in APP, which translate to amino acid positions in APP include, but are not limited to, K670N, M671L, A673V, D678H, D678N, E682K, K687N, A692G, E693G, D694N, A713T, T714A, T714I, V715A, V715M, I716F, I716M, I716T, I716V, V717F, V717G, V717I, V717L, T719P, M722K, L723P, and K724N. In some embodiments, the SNVs in APP, which translate to amino acid positions in APP include, but are not limited to, A673V, A713T, T714I, V715M, V715A, I716M, V717I, V717F, T719P, and L723P. In some embodiments, the one or more non-classical variant(s) of APP comprise a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16. - In some embodiments, the expression profile or the activity profile of an individual at risk of developing a disease or disorder characterized by unwanted accumulation of amyloid beta protein is compared to a reference expression profile or activity profile from a cohort of control individuals. In some embodiments, a presence or absence of one or more non-classical variant(s) of APP is compared to the reference expression profile or activity from the cohort of control individuals. In some embodiments, the expression profile is expression of a set of different non-classical variant(s) of APP. In some embodiments, the activity profile is activity of a set of different non-classical variant(s) of APP. In some embodiments, the non-classical variant of APP comprises a portion or all of an exon of the APP gene. In some embodiments, the non-classical variant of APP comprises a portion or all of
exon exon exon 8. In some embodiments, the non-classical variant of APP comprises a single nucleotide variation (SNV). In some embodiments, the non-classical variant of APP comprises one or more SNVs. In some embodiments, the SNV in APP, which translate to amino acid positions in APP include, but are not limited to, K670, M671, A673, D678, E682, K687, A692, E693, D694, A713, T714, V715, I716, V717, T719, M722, L723, and K724, wherein the amino acids correspond topositions 670, 671, 673, 678, 682, 687, 692, 693, 694, 713, 714, 715, 716, 717, 719, 722, 723, and 724 of SEQ ID NO: 17. In some embodiments, the SNV in APP, which translate to amino acid positions in APP include, but are not limited to, A673, A713, T714, V715, I716, V717, T719, and L723, wherein the amino acids correspond topositions 673, 713, 714, 715, 716, 717, 719, and 723 of SEQ ID NO: 17. In some embodiments, the SNV in APP, which translate to amino acid positions in APP include, but are not limited to, K670N, M671L, A673V, D678H, D678N, E682K, K687N, A692G, E693G, D694N, A713T, T714A, T714I, V715A, V715M, I716F, I716M, I716T, I716V, V717F, V717G, V717I, V717L, T719P, M722K, L723P, and K724N. In some embodiments, the SNV in APP, which translate to amino acid positions in APP include, but are not limited to, A673V, A713T, T714I, V715M, V715A, I716M, V717I, V717F, T719P, and L723P. In some embodiments, the one or more exon(s) in the one or more non-classical variant(s) of APP is rearranged as compared to control. In some embodiments, the non-classical variant of APP does not compriseexon 8 of the APP gene. In some embodiments, the non-classical variant of APP comprises a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16. - In some embodiments, the expression profile is expression level. In some embodiments, the expression level of the one or more non-classical variants is at least or about 50%, 100%, 200%, 300%, 400%, 500%, 600%, 700%, or more than 700% increased or elevated as compared to the expression level from the cohort of control individuals. In some embodiments, the activity profile is activity level. In some embodiments, the activity level of the one or more non-classical variants is at least or about 50%, 100%, 200%, 300%, 400%, 500%, 600%, 700%, or more than 700% increased or elevated as compared to the activity level from the cohort of control individuals. In some embodiments, the expression profile or activity profile is used to detect the presence of one or more non-classical variant(s) of APP.
- In some embodiments, the expression profile or the activity profile is used to diagnose an individual. In some embodiments, the individual has a disease or disorder characterized by unwanted accumulation of amyloid beta protein. In some embodiments, the individual has or is suspected of having Alzheimer's disease. In some embodiments, the individual is diagnosed with Alzheimer's disease by measuring the expression profile or the activity profile of the one or more non-classical variant(s) of APP. In some embodiments, the individual is diagnosed with Alzheimer's disease by measuring the expression profile or the activity profile of the one or more non-classical variant(s) of APP and comparing the expression profile or the activity profile to a reference expression profile or activity profile from a cohort of control individuals. In some embodiments, the individual is diagnosed with Alzheimer's disease when one or more exon(s) in the one or more non-classical variant(s) of APP is rearranged as compared to a control. In some embodiments, the expression profile is expression level. In some embodiments, the individual is diagnosed with Alzheimer's disease when the expression level is at least or about 50%, 100%, 200%, 300%, 400%, 500%, 600%, 700%, or more than 700% increased or elevated as compared to the expression level from the cohort of control individuals. In some embodiments, the activity profile is activity level. In some embodiments, the individual is diagnosed with Alzheimer's disease when the activity level is at least or about 50%, 100%, 200%, 300%, 400%, 500%, 600%, 700%, or more than 700% increased or elevated as compared to the activity level from the cohort of control individuals.
- In some embodiments, the expression profile or the activity profile is used to more accurately diagnose or treat an individual having a disease or disorder. In some embodiments, use of the expression profile or the activity profile is at least or about 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or more than 95% more accurate at diagnosing a disease or disorder. In some embodiments, use of the expression profile or the activity profile is at least or about 1.5×, 2×, 2.5×, 3×, 3.5×, 4.0×, 4.5×, 5×, 6×, 7×, 8×, 9×, 10×, or more than 10× more accurate at diagnosing a disease or disorder. In some embodiments, the disease or disorder is Alzheimer's disease. In some embodiments, methods as described herein for accurately diagnosing or treating Alzheimer's disease are improved as compared to methods comprising neurological tests, mental exams, or brain imaging (e.g. MRI, CT, or PET scans).
- In some embodiments, determining whether the individual has or is predisposed to Alzheimer's disease is based on the expression profile or the activity profile from, wherein a likelihood of having or being predisposed to Alzheimer's disease is increased when the expression profile or the activity profile is elevated compared to a reference expression profile or reference activity profile of the one or more non-classical variant(s) derived from a cohort of control individuals. Methods as described herein for determining a likelihood of having or being predisposed to Alzheimer's disease, in some embodiments, are improved as compared to methods comprising neurological tests, mental exams, or brain imaging (e.g. MRI, CT, or PET scans). In some embodiments, the likelihood of having or being predisposed to Alzheimer's disease is increased by at least or about 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or more than 95% by determining the expression profile or activity profile of the one or more non-classical variant(s). In some embodiments, the likelihood of having or being predisposed to Alzheimer's disease is increased by at least or about 1.5×, 2×, 2.5×, 3×, 3.5×, 4.0×, 4.5×, 5×, 6×, 7×, 8×, 9×, 10×, or more than 10× by determining the expression profile or activity profile of the one or more non-classical variant(s).
- In some embodiments, the expression profile or the activity profile is used for treating an individual having a disease or disorder. In some embodiments, the expression profile or the activity profile is associated with Alzheimer's disease. In some embodiments, a therapeutic agent is administered based on the expression profile or the activity profile. In some embodiments, the therapeutic agent is optimized based on the expression profile or the activity profile. In some embodiments, the expression profile or the activity profile is measured prior to a treatment, during a treatment, or after a treatment. For example the expression profile or the activity profile is measured at 1 day, 2 days, 3 days, 4 days, 5
days 6 days, 1 week, 2 weeks, 3, weeks, 4 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 1 year, 2 years, or more than 2 years before treatment. In some embodiments, the expression profile or the activity profile is measured at 1 day, 2 days, 3 days, 4 days, 5days 6 days, 1 week, 2 weeks, 3, weeks, 4 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 1 year, 2 years, or more than 2 years occurs after treatment. - Disclosed herein, in certain embodiments, are agents for treating a disease or disorder characterized by unwanted accumulation of amyloid beta protein. Further disclosed herein, in certain embodiments, are methods of treating a disease or disorder characterized by unwanted accumulation of amyloid beta protein in an individual in need thereof, comprising: administering to the individual an agent that inhibits activity of one or more non-classical variant(s) of an amyloid precursor protein (APP) gene. Further disclosed herein, in certain embodiments, are methods of diagnosing and treating a disease or disorder characterized by unwanted accumulation of amyloid beta protein in an individual in need thereof, comprising: identifying the individual as having the disease or disorder characterized by unwanted accumulation of amyloid beta protein if binding of one or more probe(s) to one or more non-classical variant(s) of an APP gene is measured; and administering to the individual having the expression profile or the activity profile of the one or more non-classical variant(s) of APP an agent that inhibits activity of the one or more non-classical variant(s) of APP. Further disclosed herein, in certain embodiments, are methods of diagnosing and treating a disease or disorder characterized by unwanted accumulation of amyloid beta protein in an individual in need thereof, comprising: identifying the individual as having the disease or disorder characterized by unwanted accumulation of amyloid beta protein if an expression profile or an activity profile of one or more non-classical variant(s) of an APP gene is measured by a method comprising long-read sequencing of a biological sample from the individual; and administering to the individual having the expression profile or the activity profile of the one or more non-classical variant(s) of APP an agent that inhibits activity of the one or more non-classical variant(s) of APP.
- In some embodiments, the agents inhibit an expression profile of the one or more non-classical variant(s) of APP. In some embodiments, the expression profile is expression level. In some embodiments, the expression profile is expression of a set of different non-classical variants. In some embodiments, the agents inhibit activity of the one or more non-classical variant(s) of APP.
- In some embodiments, the one or more non-classical variant(s) of APP comprises a portion or all of
exon exon exon 8. In some embodiments, the one or more exon(s) in the one or more non-classical variant(s) of APP is rearranged as compared to a control. In some embodiments, the one or more non-classical variant(s) of APP comprises a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16. - In some embodiments, the agents inhibit unwanted accumulation of amyloid beta protein. In some embodiments, the unwanted accumulation of amyloid beta protein is associated with a disease or disorder. In some embodiments, the disease or disorder is Alzheimer's disease. In some embodiments, the Alzheimer's disease is familial Alzheimer's disease or sporadic Alzheimer's disease. In some embodiments, the agents inhibit plaque formation comprising a portion of or all amyloid beta. In some embodiments, the agents inhibit soluble amyloid beta protein. In some embodiments, the unwanted accumulation of amyloid beta protein is associated with one or more non-classical variant(s) of APP.
- In some embodiments, the agents target one or more non-classical variant(s) of APP gene or protein thereof. Exemplary agents include, but are not limited to, an antibody, an antigen binding fragment, a RNA interfering agent (RNAi), a small interfering RNA (siRNA), a short hairpin RNA (shRNA), a microRNA (miRNA), an antisense oligonucleotide (AON), a peptide, a peptidomimetic, a small molecule, or an aptamer.
- In some embodiments, the agent is an N-methyl-D-aspartate (NMDA) receptor antagonist, or an anti-amyloid beta antibody. In some embodiments, the cholinesterase inhibitor is selected from the group consisting of Donepezil, Galantamine, and Rivastigmine. In some embodiments, the NMDA receptor antagonist is memantine. In some embodiments, the anti-amyloid beta antibody is selected from the group consisting of Bapineuzumab, Solanezumab, Gantenerumab, Crenezumab, BAN2401, Ponezumab, and Aducanumab. In some embodiments, the N-methyl-D-aspartate (NMDA) receptor antagonist, or an anti-amyloid beta antibody is administered in conjunction with an agent that targets one or more non-classical variant(s) of APP gene or protein thereof. In some embodiments, the agent is an antibody, an antigen binding fragment, a RNA interfering agent (RNAi), a small interfering RNA (siRNA), a short hairpin RNA (shRNA), a microRNA (miRNA), an antisense oligonucleotide (AON), a peptide, a peptidomimetic, a small molecule, or an aptamer that targets the one or more non-classical variant(s) of APP gene or protein thereof.
- In some embodiments, the N-methyl-D-aspartate (NMDA) receptor antagonist, or an anti-amyloid beta antibody is administered prior to administration of an agent that targets one or more non-classical variant(s) of APP gene or protein thereof. In some embodiments, the N-methyl-D-aspartate (NMDA) receptor antagonist, or an anti-amyloid beta antibody is administered up to 1 day, up to 2 days, up to 3 days, up to 5 days, or more than 5 days prior to administration of an agent that targets one or more non-classical variant(s) of APP gene or protein thereof. In some embodiments, the N-methyl-D-aspartate (NMDA) receptor antagonist, or an anti-amyloid beta antibody is administered singly, or over a time course, such as daily, multiple times weekly, weekly, biweekly, monthly or less frequently prior to administration of an agent that targets one or more non-classical variant(s) of APP gene or protein thereof.
- In some embodiments, the N-methyl-D-aspartate (NMDA) receptor antagonist, or an anti-amyloid beta antibody is administered prior to administration of an agent that targets one or more non-classical variant(s) of APP gene or protein thereof. In some embodiments, the N-methyl-D-aspartate (NMDA) receptor antagonist, or an anti-amyloid beta antibody is administered up to 1 day, up to 2 days, up to 3 days, up to 5 days, or more than 5 days prior to administration of an agent that targets one or more non-classical variant(s) of APP gene or protein thereof. In some embodiments, the N-methyl-D-aspartate (NMDA) receptor antagonist, or an anti-amyloid beta antibody is administered singly, or over a time course, such as daily, multiple times weekly, weekly, biweekly, monthly or less frequently prior to administration of an agent that targets one or more non-classical variant(s) of APP gene or protein thereof.
- In some embodiments, the agent is a small molecule. In some embodiments, the small molecule is an antagonist of APP. In some embodiments, the small molecule is an antagonist of one or more non-classical variant(s) of APP. In some embodiments, the one or more non-classical variant(s) of APP comprises a sequence as set forth in SEQ ID NO: 1, 2, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16. In some embodiments, the small molecule is an antagonist of protein encoded by the APP gene.
- In some embodiments, the agent is an antibody. Exemplary antibodies include, but are not limited to, a monoclonal antibody, a polyclonal antibody, a bi-specific antibody, a multispecific antibody, a grafted antibody, a human antibody, a humanized antibody, a synthetic antibody, a chimeric antibody, a camelized antibody, a single-chain Fvs (scFv), a single chain antibody, a Fab fragment, a F(ab′) fragment, disulfide-linked Fvs (sdFv), an intrabody, an anti-idiotypic (anti-Id) antibody, or ab antigen-binding fragments thereof. In some embodiments, the antibody comprises immunoglobulin molecules and immunologically active fragments of immunoglobulin molecules, e.g., molecules that contain an antigen binding site. Immunoglobulin molecules are of any type, class (e.g., IgG, IgE, IgM, IgD, IgA and IgY), or subclass (e.g., IgG1, IgG2, IgG3, IgG4, IgA1 and IgA2).
- In some embodiments, the antibody selectively binds to a protein encoded by one or more non-classical variant(s) of APP gene. “Selectively binds” refers to the preference of an antibody to interact with one molecule as compared to another.
- In some embodiments, the antibody specifically binds to a protein encoded by one or more non-classical variant(s) of APP gene. The phrase “specifically binds” when referring to the interaction between an antibody or other binding molecule and a protein or polypeptide or epitope, typically refers to an antibody or other binding molecule that recognizes and detectably binds with high affinity to the target of interest. Preferably, under designated or physiological conditions, the specified antibodies or binding molecules bind to a particular polypeptide, protein or epitope yet does not bind in a significant or undesirable amount to other molecules present in a biological sample. For example, the specified antibody or binding molecule does not undesirably cross-react with non-target antigens and/or epitopes.
- In some embodiments, the antibody binds to a protein encoded by the one or more non-classical variant(s) of APP. In some embodiments, the antibody binds to a protein encoded by the one or more non-classical variant(s) of APP comprising a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16. In some embodiments, the antibody does not bind to wild-type protein.
- In some embodiments, agents for inhibiting one or more non-classical variant(s) of APP comprise an antisense RNA that hybridizes to a target RNA and inhibits the activity. In some embodiments, the antisense RNA stringently hybridizes to the target RNA and inhibits the activity. In some embodiments, the target RNA is one or more non-classical variant. In some embodiments, the target RNA is one or more non-classical variant comprising a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16.
- In some embodiments, the agent is an antisense RNA molecule. Exemplary antisense RNA molecules include, but are not limited to, RNAi, siRNA, shRNA, or miRNA. In some embodiments, the antisense RNA is double stranded or single stranded. In some embodiments, the antisense RNA comprises about 1 to about 50 nucleotides. In some embodiments, the antisense RNA comprises about 5 to about, about 5 to about 30, about 10 to about 30, about 15 to about 25, or about 20 to about 25 nucleotides. In some embodiments, the antisense RNA is at least or about 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, or 100% complementary to the target RNA.
- In some embodiments, the antisense RNA inhibits activity of APP. In some embodiments, the antisense RNA is a double-stranded antisense RNA molecule (e.g., siRNA, miRNA, shRNA) that down-regulates expression of APP, wherein one of the strands of the double-stranded antisense RNA molecule comprises a nucleotide sequence that is complementary to a nucleotide sequence of APP RNA encoded by APP or a portion thereof, and wherein the second strand of the double-stranded antisense RNA molecule comprises a nucleotide sequence substantially similar to the nucleotide sequence of APP or RNA encoded by APP or a portion thereof. In some embodiments, the antisense RNA is a double-stranded antisense RNA molecule that down-regulates expression of APP, wherein each strand of the antisense RNA molecule comprises about 15 to 25, 18 to 24, or 19 to about 23 nucleotides, and wherein each strand comprises at least about 14, 17, or 19 nucleotides that are complementary to the nucleotides of the other strand. In some embodiments, the antisense RNA is a double-stranded antisense RNA molecule that down-regulates expression of APP, wherein each strand of the antisense RNA molecule comprises about 19 to about 23 nucleotides, and wherein each strand comprises at least about 19 nucleotides that are complementary to the nucleotides of the other strand. In some embodiments, the RNA interfering activity occurs within a cell. In other embodiments, the RNA interfering activity occurs in a reconstituted in vitro system.
- In some embodiments, the antisense RNA is a single-stranded antisense RNA molecule that down-regulates expression of APP, wherein the single-stranded antisense RNA molecule comprises a nucleotide sequence that is complementary to a nucleotide sequence of APP or RNA encoded by APP or a portion thereof. In some embodiments, antisense RNA is a single-stranded antisense RNA molecule that down-regulates expression of APP, wherein the antisense RNA molecule comprises about 15 to 25, 18 to 24, or 19 to about 23 nucleotides. In some embodiments, antisense RNA molecule is a single-stranded antisense RNA molecule that down-regulates expression of APP, wherein the antisense RNA molecule comprises about 19 to about 23 nucleotides. In some embodiments, the RNA interfering activity occurs within a cell. In other embodiments, the RNA interfering activity occurs in a reconstituted in vitro system.
- In some embodiments, the antisense RNA molecule is a double-stranded polynucleotide molecule comprising self-complementary sense and antisense regions, wherein the antisense region comprises a nucleotide sequence that is complementary to a nucleotide sequence in a target nucleic acid molecule or a portion thereof and the sense region has a nucleotide sequence corresponding to the target nucleic acid sequence or a portion thereof. In some embodiments, the antisense RNA molecule is assembled from two separate polynucleotides, where one strand is the sense strand and the other is the antisense strand, wherein the antisense and sense strands are self-complementary (e.g., each strand comprises a nucleotide sequence that is complementary to the nucleotide sequence in the other strand; such as where the antisense strand and sense strand form a duplex or double-stranded structure, for example wherein the double-stranded region is about 19, 20, 21, 22, 23, or more base pairs); the antisense strand comprises a nucleotide sequence that is complementary to a nucleotide sequence in a target nucleic acid molecule or a portion thereof and the sense strand comprises a nucleotide sequence corresponding to the target nucleic acid sequence or a portion thereof. In some embodiments, the antisense RNA molecule is assembled from a single oligonucleotide, where the self-complementary sense and antisense regions of the antisense RNA molecule are linked by means of a nucleic acid based or non-nucleic acid-based linker(s).
- In some embodiments, the antisense RNA molecule is a polynucleotide with a duplex, asymmetric duplex, hairpin, or asymmetric hairpin secondary structure, having self-complementary sense and antisense regions, wherein the antisense region comprises a nucleotide sequence that is complementary to a nucleotide sequence in a separate target nucleic acid molecule or a portion thereof and the sense region has a nucleotide sequence corresponding to the target nucleic acid sequence or a portion thereof. In other embodiments, the antisense RNA molecule is a circular single-stranded polynucleotide having two or more loop structures and a stem comprising self-complementary sense and antisense regions, wherein the antisense region comprises a nucleotide sequence that is complementary to a nucleotide sequence in a target nucleic acid molecule or a portion thereof and the sense region has a nucleotide sequence corresponding to the target nucleic acid sequence or a portion thereof, and wherein the circular polynucleotide is processed either in vivo or in vitro to generate an active antisense RNA molecule capable of mediating RNA interfering activity. In additional embodiments, the antisense RNA molecule also comprises a single-stranded polynucleotide having a nucleotide sequence complementary to a nucleotide sequence in a target nucleic acid molecule or a portion thereof, wherein the single stranded polynucleotide further comprises a terminal phosphate group, such as a 5′-phosphate, or 5′,3′-diphosphate.
- In some embodiments, an asymmetric duplex is a linear antisense RNA molecule comprising an antisense region, a loop portion that comprises nucleotides or non-nucleotides, and a sense region that comprises fewer nucleotides than the antisense region to the extent that the sense region has enough complimentary nucleotides to base pair with the antisense region and form a duplex with loop. For example, an asymmetric hairpin antisense RNA molecule comprises an antisense region having length sufficient to mediate RNA interfering activity in a cell or in vitro system (e.g., about 19 to about 22 nucleotides) and a loop region comprising about 4 to about 8 nucleotides, and a sense region having about 3 to about 18 nucleotides that are complementary to the antisense region. In some embodiments, the asymmetric hairpin the antisense RNA molecule also comprises a 5′-terminal phosphate group that is chemically modified. In additional embodiments, the loop portion of the asymmetric hairpin antisense RNA molecule comprises nucleotides, non-nucleotides, linker molecules, or conjugate molecules.
- In some embodiments, an asymmetric duplex is an antisense RNA molecule having two separate strands comprising a sense region and an antisense region, wherein the sense region comprises fewer nucleotides than the antisense region to the extent that the sense region has enough complimentary nucleotides to base pair with the antisense region and form a duplex. For example, an asymmetric duplex antisense RNA molecule comprises an antisense region having length sufficient to mediate RNA interfering activity in a cell or in vitro system (e.g., about 19 to about 22 nucleotides) and a sense region having about 3 to about 18 nucleotides that are complementary to the antisense region.
- In some embodiments, an antisense RNA inhibits activity of a target RNA in a cleavage-dependent process. For example, the cleavage-dependent process involves the RNA-induced silencing complex (RISC). In some embodiments, the antisense RNA (e.g., siRNA) comprises a passenger strand and guide strand. The guide strand pairs with a complementary sequence in a mRNA molecule and induces cleavage by an RNase H endonuclease of the RISC complex. In some embodiments, the RNase H endonuclease is Argonaute. In some embodiments, an antisense RNA inhibits activity in a cleavage-independent process. For example, the antisense RNA (e.g., miRNA) comprises nucleotide mismatches with their targets and effect gene silencing through translational repression of the target gene.
- In some embodiments, an antisense RNA inhibits the one or more non-classical variant(s) of APP gene. In some embodiments, the antisense RNA inhibits pre-mRNA. In some embodiments, the antisense RNA inhibits mRNA. In some embodiments, the antisense RNA alters various functions of the target RNA. In some embodiments, the antisense RNA alters splicing of the RNA to yield one or more mRNA species. In some embodiments, the antisense RNA alters translation of protein from RNA. In some embodiments, the antisense RNA alters translocation of the RNA to the site of protein translation. In some embodiments, the antisense RNA alters a catalytic activity of the RNA or which is facilitated by the RNA. Alternatively or in combination, the antisense RNA reduces an amount of pre-mRNA.
- In some embodiments, the one or more non-classical variant(s) comprise exons that are inverted. For example, the one or more non-classical variant(s) of APP comprises one or more exons of APP that are inverted. In some embodiments, the one or more non-classical variant(s) of APP comprises one or more exons of APP and one or more exons of APP that are inverted. In some embodiments, agents for inhibiting one or more non-classical variant(s) of APP, wherein the one or more exons of APP are inverted, comprise a sense RNA that hybridizes to a target RNA and inhibits the activity. In some embodiments, the sense RNA stringently hybridizes to the target RNA and inhibits the activity. In some embodiments, the target RNA is one or more non-classical variants of APP comprising inverted exons of APP.
- In some embodiments, the agent is a sense RNA molecule. In some embodiments, the sense RNA is double stranded or single stranded. In some embodiments, the sense RNA comprises about 1 to about 50 nucleotides. In some embodiments, the sense RNA comprises about 5 to about, about 5 to about 30, about 10 to about 30, about 15 to about 25, or about 20 to about 25 nucleotides. In some embodiments, the sense RNA is at least or about 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, or 100% complementary to the target RNA. In some embodiments, the target RNA comprises inverted exons of APP.
- In some embodiments, a sense RNA inhibits the one or more non-classical variant(s) of APP gene, wherein the one or more non-classical variant(s) comprises inverted exons of the APP gene. In some embodiments, the sense RNA inhibits pre-mRNA. In some embodiments, the sense RNA inhibits mRNA. In some embodiments, the sense RNA alters various functions of the target RNA. In some embodiments, the sense RNA alters splicing of the RNA to yield one or more mRNA species. In some embodiments, the sense RNA alters translation of protein from RNA. In some embodiments, the sense RNA alters translocation of the RNA to the site of protein translation. In some embodiments, the sense RNA alters a catalytic activity of the RNA or which is facilitated by the RNA. Alternatively or in combination, the sense RNA reduces an amount of pre-mRNA.
- In some embodiments, the agent is an antisense oligonucleotide (AON). In some embodiments, the AON comprises antisense oligonucleotide strands. In some embodiments, the AON comprises sense oligonucleotide strands. In some embodiments, the AON comprises antisense oligonucleotide strands and sense oligonucleotide strands. In some embodiments, the AON restores a reading frame and allow for production of functional APP. In some embodiments, the AON targets RNA of the one or more non-classical variant(s) of APP gene. In some embodiments, the RNA is pre-mRNA. In some embodiments, the RNA is mRNA. In some embodiments, the AON targets DNA of the one or more non-classical variant(s) of APP gene. In some embodiments, the DNA is genomic DNA. In some embodiments, the DNA is nuclear DNA. In some embodiments, the DNA is extrachromosomal or extranuclear DNA. In some embodiments, the DNA is circular DNA.
- Antisense oligonucleotides (AONs), in some embodiments, inhibit the expression of one or more non-classical variant(s) of APP gene. In some embodiments, the AONs inhibit the activity of the one or more non-classical variant(s) of APP gene. In some embodiments, the AONs inhibit the expression or activity of the one or more non-classical variant(s) of APP gene by targeting RNA of the one or more non-classical variant(s) of APP gene for degradation.
- In some embodiments, antisense oligonucleotides (AONs) inhibit a gene of a protein involved in transcription of APP. An exemplary protein is a transcription factor, coactivator, corepressor, chromatin modifying enzyme, histone acetyltransferase, histone deacetylase, kinase, or methylase, or any other protein involved in a signal transduction pathway that results in transcription of APP. In some embodiments, the AONs inhibit the gene of a protein involved in transcription of APP to inhibit generation of the one or more non-classical variants of APP.
- In some embodiments, the antisense oligonucleotide (AON) results in an insertion, deletion, duplication, or alteration in an incorrectly processed transcript of the APP gene. In some embodiments, the incorrectly processed transcript of the APP gene is one or more non-classical variant(s) of the APP gene. In some embodiments, the one or more non-classical variant(s) comprise a portion or all of an exon of APP. In some embodiments, the one or more non-classical variant(s) comprise a portion or all of
exon exon 8. In some embodiments, the AON induces exon skipping or exon inclusion to restore the translational reading frame of the one or more non-classical variant(s) of APP. - In some embodiments, the antisense oligonucleotide (AON) induces exon skipping. In some embodiments, the AON is a short nucleic acid sequence that binds to specific mRNA or pre-mRNA sequences of the APP gene to induce exon skipping. In some embodiments, the AON is a short nucleic acid sequence that binds to specific DNA sequences of the APP gene to induce exon skipping. In some embodiments, the AON binds splice sites or exonic enhancers. In some embodiments, binding of the AON to specific mRNA or pre-mRNA sequences generates double-stranded regions. In some embodiments, formation of double-stranded regions occurs at sites where the spliceosome or proteins associated with the spliceosome would normally bind and causes exons to be skipped. In some embodiments, skipping of exons results in restoration of the transcript reading frame and allows for production of functional APP.
- In some embodiments, the antisense oligonucleotide (AON) induces exon inclusion. In some embodiments, the AON binds to at least one of a splice site, a site near a splice site, and a site distant to a splice site. In some embodiments, the AON binds at site in the RNA to prevent disruption of an exon splice enhancer or intron splice enhancer. In some embodiments, the AON binds at site in the RNA to prevent creation of an exon splice silencer or intron splice silencer.
- In some embodiments, the antisense oligonucleotide (AON) comprises natural, synthetic, or artificial nucleotide analogues or bases. In some embodiments, the AON comprises DNA, RNA, or nucleotide analogues. In some embodiments, the synthetic or artificial nucleotide analogues or bases comprise modifications at one or more of ribose moiety, phosphate moiety, nucleoside moiety, or a combination thereof.
- In some embodiments, the antisense oligonucleotide (AON) comprises a nucleobase that is unmodified such as adenine, guanine, cytosine, thymine, and uracil or any synthetic or modified nucleobase. Examples of modified nucleobases include, without limitation, hypoxanthine, xanthine, 7-methylguanine, 5,6-dihydrouracil, 5-methylcytosine, and 5-hydroxymethoylcytosine.
- In some embodiments, the antisense oligonucleotide (AON) comprises a backbone that connects components of the AON. In some embodiments, the backbone comprises a 3′-5′ phosphodiester linkage connecting sugar moieties of the AON. Examples of a backbone structure or linkages of the AON, include, but are not limited to, phosphorothioate, phosphorodithioate, phosphoroselenoate, phosphorodiselenoate, phosphoroanilothioate, phosphoraniladate, and phosphoramidate. In some embodiments, the backbone structure of the AON does not comprise phosphorous but comprises peptide bonds, for example in a peptide nucleic acid (PNA), or linking groups including carbamate, amides, and linear and cyclic hydrocarbon groups. In some embodiments, the backbone modification is a phosphorothioate linkage. In some embodiments, the backbone modification is a phosphoramidate linkage.
- In some embodiments, the antisense oligonucleotide (AON) comprises an unmodified sugar moiety such as ribose or deoxyribose or a modified sugar moiety or sugar analog, including a morpholino ring. Non-limiting examples of modified sugar moieties include 2′ substitutions such as 2′-O-methyl (2′-O-Me), 2′-O-methoxyethyl (2′MOE), 2′-O-aminoethyl, 2′F; N3′->P5′ phosphoramidate, 2′dimethylaminooxyethoxy, 2′dimethylaminoethoxyethoxy, 2′-guanidinidium, 2′-O-guanidinium ethyl, carbamate modified sugars, and bicyclic modified sugars. In some embodiments, the sugar moiety modification is an extra bridge bond, such as in a locked nucleic acid (LNA). In some embodiments the sugar analog contains a morpholino ring, such as phosphorodiamidate morpholino (PMO). In some embodiments, the sugar moiety comprises a ribofuransyl or 2′deoxyribofuransyl modification. In some embodiments, the sugar moiety comprises 2′4′-constrained 2′O-methyloxyethyl (cMOE) modifications. In some embodiments, the sugar moiety comprises
cEt 2′, 4′ constrained 2′-O ethyl BNA modifications. In some embodiments, the sugar moiety comprises tricycloDNA (tcDNA) modifications. In some embodiments, the sugar moiety comprises ethylene nucleic acid (ENA) modifications. In some embodiments, the sugar moiety comprises MCE modifications. - In some embodiments, the antisense oligonucleotide (AON) comprises an artificial nucleotide analogue. Exemplary artificial nucleotide analogues include 2′-O-methyl, 2′-O-methoxyethyl (2′-O-MOE), 2′-O-aminopropyl, 2′-deoxy, T-deoxy-2′-fluoro, 2′-O-aminopropyl (2′-O-AP), 2′-O-dimethylaminoethyl (2′-O-DMAOE), 2′-O-dimethylaminopropyl (2′-O-DMAP), T-O-dimethylaminoethyloxyethyl (2′-O-DMAEOE), or 2′-O-N-methylacetamido (2′-O-NMA) modified, LNA, ENA, PNA, HNA, morpholino, methylphosphonate nucleotides, thiolphosphonate nucleotides, 2′-fluoro N3-P5′-phosphoramidites, or combinations thereof. In some embodiments, the modified nucleotide analogue is a constrained ethyl (cEt) nucleotide.
- In some embodiments, the antisense oligonucleotide (AON) comprises a number of nucleobases. In some embodiments, the number of nucleobases comprises a range of about 8 to 50, 8 to 40, 8 to 35, 8 to 30, 8 to 25, 8 to 20, 8 to 15, 9 to 50, 9 to 40, 9 to 35, 9 to 30, 9 to 25, 9 to 20, 9 to 15, 10 to 50, 10 to 40, 10 to 35, 10 to 30, 10 to 25, 10 to 20, 10 to 15, 11 to 50, 11 to 40, 11 to 35, 11 to 30, 11 to 25, 11 to 20, 11 to 15, 12 to 50, 12 to 40, 12 to 35, 12 to 30, 12 to 25, 12 to 20, 12 to 15, 13 to 50, 13 to 40, 13 to 35, 13 to 30, 13 to 25, 13 to 20, 14 to 50, 14 to 40, 14 to 35, 14 to 30, 14 to 25, 14 to 20, 15 to 50, 15 to 40, 15 to 35, 15 to 30, 15 to 25, 15 to 20, 20 to 50, 20 to 40, 20 to 35, 20 to 30, 20 to 25, 25 to 50, 25 to 40, 25 to 35, or 25 to 30 nucleobases.
- In some embodiments, the sequence of the antisense oligonucleotide (AON) is at least or about 40%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, or 99.5% complementary to a target sequence. In some embodiments, the target sequence is a sequence of the one or more non-classical variant(s) of APP gene. In some embodiments, the target sequence is a RNA sequence. In some embodiments, the target sequence is a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16. In some embodiments, the target sequence is a DNA sequence.
- In some embodiments, the agent inhibits transcription of APP. In some embodiments, the agent inhibits transcription of APP and subsequent incorporation of one or more non-classical variants of APP into the genome. In some embodiments, the agent inhibits a protein in a signal transduction pathway involved in the transcription of APP. In some embodiments, the protein is extracellular. Exemplary extracellular proteins include cell membrane receptors including, but not limited to, G protein-coupled receptors, integrin receptors, Notch receptors, cadherin receptors, receptor tyrosine kinase receptors, chemokine receptors, cytokine receptors, death receptors, T-cell receptors, and any combination thereof. In some embodiments, the agent targets a signaling molecule that signals through the extracellular proteins. Exemplary signaling molecules include, but are not limited to, hormones, neurotransmitters, cytokines, growth factors, cell adhesion molecules, and vitamins. In some embodiments, the agent targets a signaling molecule of an extracellular protein receptor to prevent binding of the signaling molecule and the extracellular protein to subsequently inhibit transcription of APP. In some embodiments, the agent mimics a signaling molecule of an extracellular protein receptor to inhibit signaling and subsequent transcription of APP.
- In some embodiments, the agent inhibits transcription of APP by inhibiting an intracellular protein involved in transcription of APP. In some embodiments, the protein is cytosolic. In some embodiments, the protein is nuclear. In some embodiments, the protein modulates transcription of APP. In some embodiments, the protein is a transcription factor, coactivator, corepressor, chromatin modifying enzyme, histone acetyltransferase, histone deacetylase, kinase, or methylase that modulates transcription of APP.
- Exemplary signal transduction pathways involved in transcription of APP include, but are not limited to, Wnt signal transduction pathway, 5′ adenosine monophosphate-activated protein kinase (AMPK), mechanistic target of rapamycin (mTOR) complexes, the Sirtuin 1 (silent mating-
type information regulator 2 homolog 1)/peroxisome proliferator-activated receptor gamma co-activator 1-α (Sirt1/PGC-1α) axis, and cholinergic receptor signaling. - In some embodiments, the agent edits a nucleic acid of one or more non-classical variant(s) of APP. In some embodiments, the agent edits DNA. In some embodiments, the agent edits RNA. An exemplary system for nucleic acid editing comprises Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) and a CRISPR-associated (Cas) protein. When expressed or transferred into cells alongside a guide RNA (gRNA), a Cas protein allows for the targeted introduction or deletion of genetic information via a complex with CRISPR sequence of mRNA. Generally, the gRNA comprises a target sequence region, a protospacer-adjacent motif (PAM) region, and a hairpin region. In a CRISPR/Cas process, a gRNA shepherds the Cas enzyme to a specific stretch of nucleic acid. In some embodiments, the gRNA is a single stranded guide RNA (sgRNA). In some embodiments, the gRNA is a dual stranded guide RNA (dgRNA). Cas then cleaves the nucleic acid to disable or repair a gene. In some embodiments, the nucleic acid is DNA. In some embodiments, the nucleic acid is RNA.
- Provided herein, in certain embodiments, are methods for targeting a nucleic acid of one or more non-classical variants of APP using a CRISPR/Cas system. In some embodiments, the CRISPR/Cas system targets DNA of the one or more non-classical variant(s) of APP. In some embodiments, CRISPR/Cas system targets RNA of the one or more non-classical variant(s) of APP. In some embodiments, the one or more non-classical variant(s) comprise a portion or all of an exon of APP. In some embodiments, the one or more non-classical variant(s) comprise a portion or all of
exon exon exon 8. In some embodiments, the one or more non-classical variant(s) of APP comprise a single nucleotide variation (SNV) in the APP gene. In some embodiments, the non-classical variant of APP comprises one or more SNVs in the APP gene. In some embodiments, the SNV is in the amyloid beta region of APP. In some embodiments, the SNV in APP, which translate to amino acid positions in APP include, but are not limited to, K670, M671, A673, D678, E682, K687, A692, E693, D694, A713, T714, V715, I716, V717, T719, M722, L723, and K724, wherein the amino acids correspond topositions 670, 671, 673, 678, 682, 687, 692, 693, 694, 713, 714, 715, 716, 717, 719, 722, 723, and 724 of SEQ ID NO: 17. In some embodiments, the SNV in APP, which translate to amino acid positions in APP include, but are not limited to, A673, A713, T714, V715, I716, V717, T719, and L723, wherein the amino acids correspond topositions 673, 713, 714, 715, 716, 717, 719, and 723 of SEQ ID NO: 17. In some embodiments, the SNV in APP, which translate to amino acid positions in APP include, but are not limited to, K670N, M671L, A673V, D678H, D678N, E682K, K687N, A692G, E693G, D694N, A713T, T714A, T714I, V715A, V715M, I716F, I716M, I716T, I716V, V717F, V717G, V717I, V717L, T719P, M722K, L723P, and K724N. In some embodiments, the SNV in APP, which translate to amino acid positions in APP include, but are not limited to, A673V, A713T, T714I, V715M, V715A, I716M, V717I, V717F, T719P, and L723P. - In some embodiments, a nuclease for use in the CRISPR/Cas system is from a species of, but not limited to, Streptococcus, Campylobacter, Nitratifractor, Staphylococcus, Parvibaculum, Roseburia, Neisseria, Gluconacetobacter, Azospirillum, Sphaerochaeta, Lactobacillus, Eubacterium, Corynebacter, Carnobacterium, Rhodobacter, Listeria, Paludibacter, Clostridium, Lachnospiraceae, Clostridiaridium, Leptotrichia, Francisella, Legionella, Alicyclobacillus, Methanomethyophilus, Porphyromonas, Prevotella, Bacteroidetes, Helcococcus, Letospira, Desulfovibrio, Desulfonatronum, Desulfurococcus, Opitutaceae, Tuberibacillus, Bacillus, Brevibacilus, Methylobacterium, Natronobacterium, Flavobacterium, Saccharomyces, Chlamydomonas, Thermus, Pyrococcus, Mycoplasma, or Acidaminococcus.
- Exemplary Cas proteins include, but are not limited to, Cpf1, C2c1, C2c2, Cas1, Cas1B, Cas2, Cas3, Cas4, Cas5, Cash, Cas7, Cas8, Cas9 (Csn1 or Csx12), Cas10, Cas13, Csy1, Csy2, Csy3, Cse1, Cse2, Csc1, Csc2, Csa5, Csn2, Csm2, Csm3, Csm4, Csm5, Csm6, Cmr1, Cmr3, Cmr4, Cmr5, Cmr6, Csb1, Csb2, Csb3, Csx17, Csx14, Csx10, Csx16, CsaX, Csx3, Csx1, Csx15, Csf1, Csf2, Csf3, Csf4, homologues thereof, and modified versions thereof. In some embodiments, the Cas protein targets DNA. In some embodiments, the Cas protein targets RNA. In some embodiments, the Cas protein is Cas9. In some embodiments, the Cas protein is Cas13. Cas proteins include, but are not limited to, wild-type Cas and derivatives, chimeras, or mutants thereof.
- In some embodiments, the agent modulates generation of one or more non-classical variant(s) of APP as a result of strand breaks. In some embodiments, the agent modulates generation of one or more non-classical variant(s) of APP as a result of single stranded breaks. In some embodiments, the agent modulates generation of one or more non-classical variant(s) of APP as a result of double stranded breaks (DSBs). In some embodiments, the agent inhibits formation of DSBs. In some embodiments, the agent inhibits formation of DSBs by inhibiting the cause of DSBs including, but not limited to, V(D)J recombination, class switch recombination, meiosis, ionizing radiation, oxidative free radicals, replication across a nick, and inadvertent enzyme actions.
- DSBs, in some embodiments, are repaired by a DNA repair pathway. In some embodiments, alteration in a DNA repair pathway results in an inability or reduced ability to repair DSBs. In some embodiments, agents for treating a disease or disorder characterized by unwanted accumulation of amyloid beta protein modulate a DNA repair pathway. Exemplary DNA repair pathways include, but are not limited to, non-homologous end joining (NHEJ), microhomology-mediated end joining (MMEJ), homologous recombination, mismatch repair, nucleotide excision repair, or DNA strand cross-link repair. In some embodiments, agents for treating a disease or disorder characterized by unwanted accumulation of amyloid beta protein modulate a DNA DSB repair activity.
- In some embodiments, the agent targets a gene involved in a DNA repair pathway. Exemplary genes involved in the DNA repair pathway include, but are not limited to, ATM, ATR, MRN, RAD51, BRCA1/BRCA2, KU70/80, DNA-PKcs, Artemis, Ligase IV, and XRCC4. In some embodiments, the agent targets a protein involved in the DNA repair pathway. Exemplary proteins involved in the DNA repair pathway include, but are not limited to, ATM, ATR, CHK1, RAD51, RAD54, PARP1, ERCC1, DNA-PKcs, and Ligase IV.
- In some embodiments, the agent inhibits a protein involved in a DNA repair pathway, wherein the DNA repair pathway is non-homologous end joining (NHEJ). In some embodiments, the protein is involved in a step of NHEJ. For example, the protein is involved in DNA termini recognition, bridging of the DNA ends, DNA end processing, or DNA recognition. Exemplary proteins involved in NHEJ include, but are not limited to, DNA-PKcs, KU70/80, Artemis, Ligase IV/XRCC4, Pol μ, or Pol λ. In some embodiments, the agent is an inhibitor of a protein involved in NHEJ. Exemplary inhibitors of DNA-PKcs include, but are not limited to, wortmannin, LY294002, NU7026, NU7441, KU-0060648, MSC2490484A, CC-122, and CC-115. Exemplary inhibitors of Ligase IV include, but are not limited to, L189 and SCR7.
- In some embodiments, the agent inhibits a protein involved in a DNA repair pathway, wherein the DNA repair pathway is homologous recombination. In some embodiments, the agent inhibits CHK1, MRE1, RAD51, or RAD54. In some embodiments, the inhibitor of CHK1 is UCN-01. In some embodiments, the inhibitor of MRE11 is mirin. In some embodiments, the inhibitor of RAD51 is RI-1 or RI-2. In some embodiments, the inhibitor of RAD54 is streptonigrin.
- In some embodiments, the agent inhibits a protein involved in one or more DNA repair pathways. For example, the inhibitor inhibits a protein involved in homologous recombination and non-homologous end joining (NHEJ). Exemplary proteins involved in homologous recombination and NHEJ include, but are not limited to, ATM and ATR. In some embodiments, the inhibitor of ATM is KU55933. In some embodiments, the inhibitor of ATR is caffeine, VE-821, or NU6027.
- In some embodiments, the agent inhibits a protein involved in a DNA repair pathway, wherein the DNA repair pathway comprises repair of single stranded breaks. In some embodiments, the agent inhibits PARP1. Exemplary PARP1 inhibitors include, but are not limited to, Olaparib (AZD2281), Iniparib (BSI 201), Rucaparib (AG014699), Velparib (ABT-888), Talazoparib (BMN-673), CEP 9722, MK 4827, BMN-673, NU1025, E7016, BGB-290, and 3-aminobenzamide.
- Methods for Screening
- Described herein, in certain embodiments, are methods for screening for therapeutic agents for treating a disease or disorder in an individual characterized by unwanted accumulation of amyloid beta protein. Further provided herein, in certain embodiments, are in vitro methods for screening for a therapeutic agent for treating a disease or disorder characterized by unwanted accumulation of amyloid beta protein, comprising: contacting a cell that expresses a non-classical variant of an amyloid precursor protein (APP) gene with a test agent; detecting inhibition of expression of the non-classical variant of the APP gene compared to a control; and identifying the test agent as a therapeutic agent if the test agent inhibits expression of the non-classical variant of the APP gene compared to the control. Further provided herein, in certain embodiments, are in vitro methods of screening for a therapeutic agent for treating a disease or disorder characterized by unwanted accumulation of amyloid beta protein, comprising: contacting a cell that expresses a non-classical variant of an APP gene with a test agent; detecting inhibition of the activity of the non-classical variant of the APP gene as compared to a control; and identifying the test agent as the therapeutic agent if the test agents inhibits activity of the non-classical variant of an APP gene as compared to the control. Further provided herein, in certain embodiments, are in vitro methods of screening for a therapeutic agent for treating a disease or disorder characterized by unwanted accumulation of amyloid beta protein, comprising: contacting a cell that expresses a non-classical variant of an APP gene with a test agent; detecting binding of the test agent to the non-classical variant of the APP gene; and identifying the test agent as the therapeutic agent if the test agents binds to the non-classical variant of the APP gene.
- In some embodiments, the therapeutic agents are screened using various methods known in the art. For example, the one or more non-classical variant(s) of APP are expressed (e.g., by transfection or transduction) in a cell or organism, contacted with the therapeutic agents, and assayed for changes in activity. In some embodiments, the cell is a neuron. In some embodiments, the therapeutic agents are assayed for binding, specificity, stability, or downstream activity.
- In some embodiments, the therapeutic agents are screened for inhibition of expression of the non-classical variant of APP gene. In some embodiments, expression of the non-classical variant of APP is measured by qPCR or gel electrophoresis. In some embodiments, inhibition of protein expression of a protein encoded by the non-classical variant of the APP gene is measured. Exemplary methods for measuring protein expression include, but are not limited to, Western blot, enzyme-linked immunosorbent assays (ELISA), or chromatography. Example of chromatography methods, include but are not limited to, high-performance liquid chromatography (HPLC) or liquid chromatography-mass spectrometry (LC/MS).
- In some embodiments, the therapeutic agents are screened for inhibition of activity of the non-classical variant of APP. For example, the activity of the non-classical variant comprises accumulation of amyloid beta protein. In some embodiments, the accumulation of amyloid beta protein is measured by a method comprising Western blot, enzyme-linked immunosorbent assays (ELISA), or chromatography. In some embodiments, the therapeutic agents are screened using an enzymatic activity assay or reporter protein activity assay. For example, the one or more non-classical variant(s) are engineered to express a reporter gene. Exemplary reporter genes include, but are not limited to, acetohydroxyacid synthase (AHAS), alkaline phosphatase (AP), beta galactosidase (LacZ), beta glucuronidase (GUS), chloramphenicol acetyltransferase (CAT), green fluorescent protein (GFP), red fluorescent protein (RFP), yellow fluorescent protein (YFP), cyan fluorescent protein (CFP), horseradish peroxidase (HRP), luciferase (Luc), nopaline synthase (NOS), octopine synthase (OCS), luciferase, and derivatives thereof. Methods to determine modulation of a reporter gene are well known in the art, and include, but are not limited to, fluorometric methods (e.g. fluorescence spectroscopy, Fluorescence Activated Cell Sorting (FACS), fluorescence microscopy), and antibiotic resistance determination.
- In some embodiments, therapeutic agents are screened for reducing activity or expression of one or more non-classical variant(s) of APP or protein thereof. In some embodiments, the therapeutic agents are identified as therapeutic agents if activity is reduced by at least or about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or more than 90%. In some embodiments, the therapeutic agents are identified as therapeutic agents if expression is reduced by at least or about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or more than 90%. In some embodiments, the therapeutic agents are identified as therapeutic agents if accumulation of amyloid beta protein is by at least or about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or more than 90%.
- Therapeutic agents, in some embodiments, are identified by binding assays. Exemplary binding assays, include but are not limited to, radioactive binding assays, fluorescence resonance energy transfer, surface plasmon resonance, enzyme-linked immunosorbent assays (ELISA), kinetic exclusion assays, and crystallography assays.
- In certain embodiments, following identification of therapeutic agents, the therapeutic agents are used for treating an individual in need thereof. Described herein, in certain embodiments, are methods of treating a disease or disorder characterized by unwanted accumulation of amyloid beta protein in an individual in need thereof, comprising: administering to the individual an agent that inhibits activity of one or more non-classical variant(s) of an APP gene. In some embodiments, the one or more non-classical variant(s) comprises a portion or all of
exon exon exon 8. In some embodiments, the one or more exon(s) in the one or more non-classical variant(s) of APP is rearranged as compared to a control. In some embodiments, the one or more non-classical variant(s) of APP comprises a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16. In some embodiments, the disease or disorder is Alzheimer's disease. In some embodiments, the Alzheimer's disease is familial Alzheimer's disease (FAD) or sporadic Alzheimer's disease. In some embodiments, the agent that inhibits the activity of APP is an antibody, an antigen binding fragment, a RNA interfering agent (RNAi), a small interfering RNA (siRNA), a short hairpin RNA (shRNA), a microRNA (miRNA), an antisense oligonucleotide, a peptide, a peptidomimetic, a small molecule, or an aptamer. In some embodiments, the antibody binds to a protein encoded by the one or more non-classical variant(s) of APP. In some embodiments, the protein is encoded by the one or more non-classical variant(s) of APP comprising a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16. - Described herein are kits for identifying one or more non-classical variant(s) of amyloid precursor protein (APP) gene. In some embodiments, kits are provided for detecting a portion or all of
exon exon exon 1 andexon 11,exon 1 andexon 14,exon 2 andexon 17,exon 2 andexon 14,exon 2 andexon 18,exon 2 andexon 16,exon 3 andexon 16,exon 3 andexon 14,exon 3 andexon 17,exon 6 andexon 17,exon 6 andexon 18,exon 3 andexon 9,exon 2 andexon 9,exon 16 andexon 18,exon 6 andexon 12,exon 5 andexon 16, orexon 16 andexon 17. In some embodiments, kits are provided for detecting the one or more non-classical variant(s) of APP that comprise a sequence as set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16. - In some embodiments, kits comprise nucleic acid or polypeptide isolation reagents. In some embodiments, kits comprise one or more probe(s) for hybridization or amplification of a target nucleic acid whose expression profile or activity profile is associated with Alzheimer's disease. In some embodiments, kits include one or more probe(s) for control genes, such as housekeeping genes. In some embodiments, the one or more probe(s) for control genes are used, for example, in ΔCt calculations. In some embodiments, a probe of the one or more probe(s) is labeled with an enzyme, a radioactive isotope, or a fluorescent label. In some embodiments, the probe is labeled using an affinity tag. Exemplary affinity tags include, but are not limited to, biotin, desthiobiotin, histidine, polyhistidine, myc, hemagglutinin (HA), FLAG, glutathione S transferase (GST), or derivatives thereof. In some embodiments, the affinity tag is recognized by avidin, streptavidin, nickel, or glutathione. In some embodiments, the kit comprises a detecting reagent that binds to the one or more probe(s). In some embodiments, the detecting reagent comprises a radioactive isotope or a fluorescent label
- In some embodiments, kits include a carrier, package, or container that is compartmentalized to receive one or more containers such as vials, tubes, and the like, each of the container(s) including one of the separate elements to be used in a method described herein. Suitable containers include, for example, bottles, vials, syringes, and test tubes. In other embodiments, the containers are formed from a variety of materials such as glass or plastic.
- In some embodiments, kits comprise one or more additional containers, each with one or more of various materials (such as reagents, optionally in concentrated form, and/or devices) desirable from a commercial and user standpoint for use of described herein. Non-limiting examples of such materials include, but not limited to, buffers, primers, enzymes, diluents, filters, carrier, package, container, vial and/or tube labels listing contents and/or instructions for use and package inserts with instructions for use. A set of instructions is optionally included. In some embodiments, a label is on or associated with the container. In some embodiments, a label is on a container when letters, numbers or other characters forming the label are attached, molded or etched into the container itself; a label is associated with a container when it is present within a receptacle or carrier that also holds the container, e.g., as a package insert. In some embodiments, a label is used to indicate that the contents are to be used for a specific therapeutic application. In some embodiments, a label also indicates directions for use of the contents, such as in the methods described herein.
- The following examples are given for the purpose of illustrating various embodiments of the invention and are not meant to limit the present invention in any fashion. The present examples, along with the methods described herein are presently representative of preferred embodiments, are exemplary, and are not intended as limitations on the scope of the invention. Changes therein and other uses which are encompassed within the spirit of the invention as defined by the scope of the claims will occur to those skilled in the art.
- Non-classical variants were identified from neurons isolated from non-diseased brains and Alzheimer's disease brains.
- Fluorescence Activated Cell Sorting of Neurons
- Neuronal nuclei were isolated from postmortem frontal cortices (CTX) and cerebellums (CBL) of non-diseased (Non-AD) and Alzheimer's disease (AD) brains and prepared for fluorescence activated cell sorting (FACS). Isolated nuclei were fixed and labeled with rabbit anti-NeuN antibody (1:800) (Millipore, Germany) and Alexa Fluor 488 donkey anti-rabbit IgG secondary (1:500) (Life Technologies, Carlsbad, Calif.), and counterstained with propidium iodide, PI (50 μg/ml) (Sigma, St. Louis, Mo.). Electronically gated diploid neuronal nuclei, determined by PI fluorescence and immunolabeling, were analyzed and sorted.
- RNA Extraction
- Following FACS, RNA was extracted from populations of 50 NeuN positive nuclei. Extracted RNA were reverse-transcribed with (CTAGTTCTGCATCTGCTCAAAGAACTTG) (SEQ ID NO: 18) and amyloid precursor protein (APP) cDNA was amplified by polymerase-chain reactions (PCR) using a forward primer (ATGCTGCCCGGTTTGGCA)(SEQ ID NO: 19) and a reverse primer (CTAGTTCTGCATCTGCTCAAAGAACTTG) (SEQ ID NO: 20).
- Half of the PCR products were run and separated on agarose gels with a DNA ladder (M) (
FIG. 2 ). Gels were transferred to nylon membranes and then blotted with P32-labelled APP cDNA probe (SEQ ID NO. 21) as seen in Table 3. Radioactivity on the membranes was detected by Typhoon phosphorimager (FIG. 3 ). -
TABLE 3 SEQ ID NO Name Sequence 21 APP ACTGCTCCTGCTGGCCGCCTGGACGGCTCGGGCGCTG cDNA GAGGTACCCACTGATGGTAATGCTGGCCTGCTGGCTG Probe AACCCCAGATTGCCATGTTCTGTGGCAGACTGAACAT GCACATGAATGTCCAGAATGGGAAGTGGGATTCAGAT CCATCAGGGACCAAAACCTGCATTGATACCAAGGAAG GCATCCTGCAGTATTGCCAAGAAGTCTACCCTGAACT GCAGATCACCAATGTGGTAGAAGCCAACCAACCAGTG ACCATCCAGAACTGGTGCAAGCGGGGCCGCAAGCAGT GCAAGACCCATCCC - The remaining PCR products were run on another agarose gel, and the bands corresponding to the positive signals (indicated by arrows) on nylon membranes were excised (
FIG. 4 ). Excised PCR products were cloned and sequenced for variant analysis. Following sequencing, non-classical variants as seenFIG. 5 were identified. - A cDNA library was prepared from populations of 50-nuclei from non-diseased (Non-AD) and Alzheimer's disease (AD) brains with lambda phage library system (Clonetech Laboratories).
- Percentages of non-classical variants cAPP-R3/16, ncAPP-R2/17, ncAPP-R1/13, and cAPP-R1/11 were determined in Non-AD and AD nuclei as seen in Table 4.
-
TABLE 4 Non-classical Variant Non-AD AD cAPP-R3/16 25% 89.4% ncAPP-R2/17 75% 0% ncAPP-R1/13 0% 5.3% cAPP-R1/11 0% 5.3% - Percentages of amyloid beta positive phage clones from Non-AD and AD brains were also determined using probes for amyloid beta (GATGCAGAATTCCGACATGACTCAGGATATGAAGTTCATCATCAAAAATTGGTG TTCTTTGCAGAAGATGTGGGTTCAAACAAAGGTGCAATCATTGGACTCATGGTGG GCGGTGTTGTCATAGCG) (SEQ ID NO: 22) and APP cDNA (
FIG. 6A ). A significant increase in percentage of amyloid beta was seen in AD brains than non-AD brains (FIG. 6B ). - Comparing APP mRNA in sorted nuclei or bulk RNA,
FIG. 7 shows that full-length APP mRNA is mainly in the cytoplasm. - Neuronal nuclei were sorted from human postmortem frontal cortices from non-diseased and Alzheimer's disease brains. Genomic DNA was extracted and purified using DNeasy Blood and Tissue Kit (Qiagen, Valencia, Calif.). Purified genomic DNA was used as a template for PCR amplification using primers for APP comprising a forward primer (ATGCTGCCCGGTTTGGCA) (SEQ ID NO: 23) and a reverse primer (CTAGTTCTGCATCTGCTCAAAGAACTTG) (SEQ ID NO: 24). PCR products amplified with APP primers were run on an agarose gel (
FIG. 8A ) as well as a no template control (NTC). In addition to APP, PSEN1 primers were used to amplify the purified genomic DNA and were run on an agarose gel with a positive control (PC) and a no template control (NTC) (FIG. 8B ). PCR products were cloned and sequenced for variant identification. - Referring to
FIG. 8A , non-classical variants of APP were identified in neuronal and non-neuronal gDNA. Using primers for PSEN1, non-classical variants were not identified on gDNA (FIG. 8B ). - Populations of 20-nuclei were isolated as in Example 1 and analyzed by quantitative polymerase chain reaction (qPCR) to detect non-classical variant cAPP-R3/16.
- Genomic DNA from sorted nuclei was extracted by QuickExtract DNA extraction solution (Epicentre) and preamplified by TaqMan PreAmp Master Mix (Thermo Fisher Scientific). Standard qPCR reactions using TaqMan probe based assays were performed in triplicate. Reactions were run on a BioRad qPCR thermocycler using TaqMan Real-Time PCR Master Mix (Thermo Fisher Scientific). The crossing threshold (Ct) was determined for primers for cAPP-R3/16, PSEN1 and TERT within the linear region of the amplification curve.
- Referring to
FIG. 9 , the non-classical variant cAPP-R3/16 was quantitatively detected in neuronal and non-neuronal cells with a wide range of distribution whereas as PSEN1 was with a consistent distribution. - Non-classical variants were detected using a DNA pull-down assay. The in vitro transcribed RNA probe sequence used for DNA pull-down was APP cDNA sequence. The pull-down sequences were cloned and sequenced for APP variant analysis.
- Neuronal nuclei were isolated from frontal cortices of AD brains as described in Example 1 and analyzed for intron/exon and exon/exon sequences of APP.
- gDNA in nuclei were hybridized with intron/exon and exon/exon probes and labeled with different colors by chromogenic method. Briefly, neuronal nuclei from AD brains were fixed and sorted for NeuN positivity, and dried onto slides. Neuronal nuclei were treated with RNase cocktail (Ambion) for 1 hour at 40° C., following by hydrogen peroxide treatment for 10 minutes at room temperature and protease treatment for 10 minutes at 40° C. DNA denaturation was performed by incubating the slides with 0.58×SSC, 70% formamide, and 0.1% SDS for 20 minutes at 80° C. DNA in situ hybridization probes were incubated with samples at 40° C. overnight.
- Chromogenic developing procedures were performed according to manufacturer's protocol (Advanced Cell Diagnostics). Slides were then visualized by microscopy.
- As seen in
FIG. 11 , genomic rearrangements were observed. Specifically, exon-exon junctions as seen in the arrow were present. - Brain samples from patients with AD were isolated and sectioned, and non-classical variants were analyzed.
- Non-classical variants were analyzed using RNA in situ hybridization. 10 μm human AD frontal cortices were sectioned and fixed by neutral buffered formalin. Fixed tissue sections were treated with hydrogen peroxide for 10 minutes at room temperature, followed by target retrieval and protease treatment. RNA in situ hybridization probes were incubated with samples for 2 hours at 40° C. Chromogenic developing procedures were performed according to manufacturer's protocol. After RNA in situ hybridization, anti-A13 monoclonal antibody (MOAB, Millipore) was incubated with sample at room temperature overnight. Horseradish peroxidase based developing method was used for signal detection. Slides were then visualized by microscopy at 200× and 630× magnification.
- As seen in
FIG. 12 , intraexonic rearrangement junctions were observed. Specifically, the non-classical variant cAPP-R3/16 (red, 1201) was detected near amyloid beta plaques (brown, 1203). - Non-classical variants were detected by single molecule real-time (SMRT) sequencing.
- RNA Samples were prepared from 2 AD temporal lobes. Methods for preparation of RNA for sequencing were provided according to manufacturer's instructions (Pacific Biosciences). Briefly, target cDNA was prepared and captured by xGene lockdown probes. cDNA of interest was then ligated to adaptors and ready for SMRT sequencing with RSII sequencer.
- Following cDNA sequencing, non-classical variants were identified (
FIG. 13 ). - Non-neuronal and neuronal nuclei were isolated from non-diseased and AD brains as described in Example 1 and analyzed for exon-exon junctions and intraexonic rearrangements of APP. Non-classical variants were analyzed using DNA in situ hybridization. Briefly, nuclei dried on to slides were treated with RNase cocktail for 1 hour at 40° C., followed by hydrogen peroxidase treatment, target retrieval, protease treatment, and DNA denaturation. DNA in situ hybridization probes were incubated with samples at 40° C. overnight. Chromogenic developing procedures were performed according to manufacturer's protocol. Probes used here were designed to detect
exon 16 andexon 17. - Nuclei from neuronal and non-neuronal cells showed an increased signal in AD brains as compared to non-diseased brains (
FIG. 14 ). - Non-classical variants were analyzed for ability to express protein.
- Non-classical variants cAPP-R3/16, cAPP-R2/16, and ncAPP-R1/4 were epitope tagged with hemagglutinin (HA). The non-classical variants were transfected and expressed in cells. Referring to
FIG. 15A , nuclei were stained with DAPI (right panel). The epitope tagged non-classical variants also heterologously expressed EGFP (center panel). Non-classical variants comprising coding regions expressed HA-tagged protein (left panel). Western blot for HA showed similar results in that non-classical variants comprising coding regions expressed HA-tagged protein (FIG. 15B ). - LN-229 cells were transfected with vehicle, non-classical variant cAPP-R3/16, or wild-type APP cDNA (APP). RNA was extracted from the LN-229 cells and subject to reverse transcription PCR (RT-PCR). RT-PCR products were run on a gel. The non-classical variant cAPP-R3/16 and APP were detected (
FIG. 16A ). - LN-229 cells were also transfected with vehicle, non-classical variant cAPP-R3/16, or APP for analysis by RNA in situ hybridization. Cells were fixed in neutral buffered formalin for 10 min at room temperature. Fixed cells were treated with hydrogen peroxide, following by target retrieval and protease treatment. RNA in situ hybridization probes were incubated with samples for 2 hours at 40° C. Chromogenic developing procedures were performed according to manufacturer's protocol. Probes used for staining were cAPP-R3/16 and PPM as a positive control. The non-classical variant cAPP-R3/16 was detected in cells expressing non-classical variant cAPP-R3/16 as well as in cells expressing wild-type APP (
FIG. 16B ). - Nuclei Extraction and Fluorescence-Activated Nuclear Sorting (FANS)
- For in situ, hybridization analyses, isolated nuclei were fixed in 1:10 diluted buffered formalin (Fisher Healthcare) for 5 minutes. Fixed or unfixed nuclei were then labeled with anti-NeuN rabbit monoclonal antibody (1:800) (Millipore, Germany) and Alexa Fluor 488 donkey anti-rabbit IgG (1:500) (Life Technology, Carlsbad, Calif.), and counterstained with propidium iodide (PI) (50μ/ml) (Sigma, St. Louis, Mo.). Diploid NeuN positive and negative nuclei were gated by PI and immunofluorescence, and sorted into appropriate populations for RT-PCR or genomic DNA PCR and in situ hybridization. FANS was performed by FACS-Aria with a FACS-Aria II.
- RNA Extraction and RT-PCR
- RNA extraction from 50-nuclei populations and bulk tissues were performed using Quick-RNA MicroPrep (Zymo Research, Irvine, Calif.) and RNAeasy Mini kits (Qiagen, Valencia, Calif.), respectively, according to manufacturer's protocol. OneStep Ahead RT-PCR (Qiagen, Valencia, Calif.) was used for RT-PCR with
APP sense primer 5′-ATGCTGCCCGGTTTGGCA-3′ (SEQ ID NO: 25) and APPanti-sense primer 5′-CTAGTTCTGCATCTGCTCAAAGAACTTG-3′ (SEQ ID NO: 26). Low annealing stringency PCR was carried out with the following thermal cycling steps: 95° C. 15 seconds, 55° C. 15 seconds, and 68° C. 2.5 minutes. - Southern Blotting
- RT-PCR products were run on an agarose gel, denatured, and transferred to a positively charged nylon membrane. UV crosslinked membranes were incubated with denatured and purified 32P-labelled APP cDNA probes at 42° C. overnight. Blots were washed four times with increasing washing stringency. Images were developed by Typhoon (GE Healthcare Life Sciences) or Fujifilm FLA-5100 phosphorimager.
- DNA Extraction and Genomic DNA PCR
- DNA extraction from isolated neuronal nuclei populations was performed using DNAeasy and QIAamp DNA Mini kits (Qiagen, Valencia, Calif.) according to manufacturer's instruction. High annealing stringency PCR for APP was performed by FastStart PCR master (Sigma, St. Louis, Mo.) with 95° C. 30 seconds, 65° C. 30 seconds, and 72° C. 2.5 minutes, and Platinum SuperFi DNA polymerase (Life Technology) with 98° C. 10 seconds, 65° C. 10 seconds, and 72° C. 1.5 minutes. For PSEN1 PCR, the primer sequences were the following:
sense 5′-ATGACAGAGTTACCTGCACC-3′ (SEQ ID NO: 27) andanti-sense 5′-CTAGATATAAAATTGATGGAA-3′ (SEQ ID NO: 28). Thermal cycling steps were 95° C. 30 seconds, 52° C. 30 seconds, 72° C. 2 minutes, and 98° C. 10 seconds, 52° C. 10 seconds, 72° C. 1 minute for FastStart PCR master and Platinum SuperFi DNA polymerase, respectively. - Junction-Specific Genomic In Situ Hybridization (jgISH) and RNA-ISH
- For jgISH pretreatment, sorted nuclei were dried on Plus Gold slides (Fisher Scientific, Pittsburgh, Pa.). Nuclei were then treated with RNase cocktail enzyme mix (1:50) (ThermoFisher) at 40° C. for 60 minutes, followed by 1:10 dilution buffered formalin fixation at room temperature for 5 minutes. After two washes with distilled water, slides were treated with hydrogen peroxide at room temperature for 10 minutes, target retrieval reagent at 95° C. for 15 minutes, followed by protease treatment at 40° C. for 10 minutes. Restriction enzyme was applied after protease treatment for 2 hours if needed. DNA was denatured (2×SSC, 70% formamide and 0.1% sodium dodecyl sulfate) at 80° C. for 20 minutes. After cooling down the slides to room temperature, probes were applied and incubated with nuclei at 40° C. overnight. Samples were then developed.
- For RNA-ISH pretreatment, 10 μm fresh frozen human tissue sections were fixed by 1:10 dilution buffered formalin on ice for 10 minutes. After two washes with PBS, tissue sections were soaked in serial diluted ethanol (50%, 70% and 100%) for 5 minutes at each step. Slides were then treated with hydrogen peroxide at room temperature for 10 minutes, followed by protease at room temperature for 20 minutes. Probes were incubated with tissue sections at 40° C. for 2 hours. Hydrogen peroxide, 10× target retrieval buffer, proteases, probes (Ex16/17 targeting ACATGACTCAGGATATGAAGTTCATCATCAAAAATTGGTGT TCTTTGCA (SEQ ID NO: 29);
IEJ 3/16 targeting TGCCAAGAAGTCTACCCTGAACTGCAGATCACCAAGATGGATGC (SEQ ID NO: 30, including sense and anti-sense probes) and reagents for signal developing were all purchased from Advanced Cell Diagnosis (ACD, Newark, Calif.). Nuclei or tissue sections were counterstained with hematoxylin. Zeiss AX10 Imager.M2 microscope and ZEN2 software were used for image acquisition. Images were thresholded, and foci number/size were quantified using ImageJ for statistical analysis. - SMRT Sequencing
- Neuronal DNA was used as template for APP PCR by Platinum SuperFi DNA polymerase with high annealing stringency (98° C. 10 seconds, 65° C. 10 seconds, and 72° C. 1.5 minutes). Multiple PCR reactions were pooled and purified by DNA Clean and Concentrator-5 (Zymo Research, Irvine, Calif.) for SMRT sequencing library preparation. PCR products were repaired using SMRTbell template prep kit version 2.0 (PacBio) and purified using AMPure PB beads (PacBio). Adapters were ligated to DNA to create SMRTbell libraries. Sequencing polymerase was annealed, and the SMRTbell library was loaded using Magbead binding. Raw bam sequencing files were converted to fastq format using the ccs2 algorithm in SMRTLink Version 4.0. Reads were only included in the analyzed fastq file if 1) there were more than 20 passes of the sequencing polymerase over the DNA molecule in the zero mode waveguide well and 2) the read was calculated to possess a >0.9999 predicted accuracy.
- Genomic Data Analyses with Customized Bioinformatic Algorithms
- Novel algorithms were developed to detect and analyze exon rearrangement in genes of interest. The algorithms were specifically designed to analyze long-read sequences generated by Pacific Biosciences Sequel platform. A series of quality control (QC) procedures were performed prior to sequence processing to ensure high quality of reads being analyzed.
- Quality Control: Consensus Sequence and Read Quality.
- PacBio circular consensus sequences (CCS) reads with less than 20 passes were filtered out to ensure overall sequence quality. Quality score and read length distributions are examined: for APP gene PCR enriched sequences, average median read-wide Phred score is 93 and read length ranged from 64 to 2470 nucleotides. Reads for which the median Phred score was >85 were analyzed.
- Quality Control: Sequencing Artifacts.
- Errors in homopolymers were handled with a method combining quality score information and reference sequence at the beginning of a homopolymer. The CCS FASTQ files encoded uncertainty in the homopolymer run length in the first Phred score of each run. If this Phred score was lower than a threshold of 30, then this position was marked as a likely sequencing artifact and not a real variant.
- PCR Primer Filter.
- The reads were checked to ensure the correct start and end sites with forward and reverse PCR primer sequences. BLAST (command line tool “blastn” 2.6.0+) was used to align primer sequences in either orientation to each read with
word size 13, gapopen penalty 0 andgap extension penalty 2. Any read where both primers were not detected was filtered out. Furthermore, reads on the negative strand were reverse complemented in this step. BLAST seed length was optimized to avoid ambiguity and ensure sensitivity. - Alignment to APP Reference Sequences.
- Ensembl reference sequence for APP protein was downloaded from the GRCh38 reference human genome assembly using the UCSC Genome Browser (http://genome.ucsc.edu/cgi-bin/hgGateway) with RefSeq accession number NM_000484.3. Since the PCR primers started at the start codon and end with the stop codon, sequences of
exons outfmt 6, -wordsize 25, -gapopen 0, -gapextend 2. These resulting alignment coordinates were used to mark regions of each read covered by exons for analysis of exon arrangements, lengths and patterns of exon-exon joins. - Construction and Retroviral Transduction of
Human APP Exon 16/Exon 17 Concatamers. - Phosphorylated oligonucleotides (Integrated DNA Technologies) composed of
human APP exon 16 andexon 17 sequences with BamHI and BglII restriction sites on the 5′ ends were annealed, and ligated into the BamHI site of the retroviral expression vector S-003-AB LZRSpBMN-linker-IRES-EGFP. Single and concatamerized oligonucleotide inserts were identified by PCR using primers flanking the BamHI insertion site and identified clones were sequenced to confirm insert copy number (GENEWIZ). Helper-free ecotropic virus was produced by transfecting DNA constructs (Lipofecatime 2000, Thermo Fisher Scientific) with single or multiple copies of the oligonucleotide inserts into the retrovirus packaging line Phoenix-ECO. 48 hours post-transfection, retroviral supernatants were harvested and 2 mL of selected virus was used for transduction of NIH-3T3 cells in 6 well plates. Retroviral transduction was carried out by removing the cell growth medium, replacing it with 2 mL of retroviral supernatant containing 4 μg/ml polybrene, and spinning at 25° C. for 1 hour at 2800 r.p.m. 48 hours post-transduction, the percentage of GFP+ cells, as identified by flow cytometry, was used to evaluate the transduction efficiency. The following primers were used to produce the retroviral constructs: 16/17 Bam: 5′-GATCCACATGACTCAGGATATGAAGTTCATCATCAAAAATTGGTGTTCTTTGCAA-3′, (SEQ ID NO: 31) and 16/17 BglII Rev: 5′-GATCTTGCAAAGAACACCAATTTTTGATGATGAACTTCATATCCTGAGTCATGTG-3′ (SEQ ID NO: 32). - Cell Culture
- NIH-3T3 cells were purchased from ATCC. Cells were maintained in Dulbecco's modified Eagle's medium (Invitrogen) containing 5% fetal bovine serum (Invitrogen) at 37° C. under 5% CO2.
- Non-classical variants were analyzed in transcriptionally amplified RNA from populations of neuronal nuclei.
- Non-classical variant sequences of APP were analyzed by RT-PCR in nuclei isolated by fluorescence activated nuclear sorting (FANS). The workflow (
FIG. 17A ) commenced with FANS to isolate neurons from both non-diseased and verified SAD prefrontal cerebral cortex (Table 5), which were run in parallel. Groups of 50, NeuN-positive neuronal nuclei were isolated and processed for RT-PCR (FIG. 17A ). Validated primers capable of amplifying full-length APP cDNA (APP 770, NM_000484.3) were used, followed by agarose gel electrophoresis. -
TABLE 5 Brain Information. Brain PMI Age Name Braak Sex (Hours) (years) SAD-1 6 F 6 88 SAD-2 6 F 12 88 SAD-3 6 F 6 84 SAD-4 6 F 4 86 SAD-5 6 M 5 83 SAD-6 6 F 10 72 ND-1 1 M U 87 ND-2 1 F 72 83 ND-3 U M U 83 ND-4 1 F 12 80 ND-3 1 F 18 93 ND-6 2 M 12 94 ND-7 U M 12 69 SAD-7 5 F 3.7 77 F = Female, M = Male, U = Unknown - In small population RT-PCR, the splice variants APP 751 (NM_201413.2) and APP 695 (NM_201414.2) were detected. Smaller bands of varied sizes were also detected (
FIG. 17B ). RT-PCR on bulk RNA detected the highly expressedcanonical APP 751 splice variants as the major product as well as smaller bands (FIG. 17C ). These RT-PCR products were Southern blotted with 32P-labeled APP cDNA probes (FIG. 17D ), which produced positive bands from duplicate gels, that were cloned and Sanger sequenced.APP splice variants FIG. 17E ). - Twelve non-classical variant sequences with IEJs were identified (
FIG. 17F ). One non-classical variant sequence was characterized by an IEJ between the 24th nucleotide ofexon 3 and 45th nucleotide of exon 16 (FIG. 17F , “R3/16”). The sequence complementarity of joined exons was found in 11 IEJs ranging in overlap from 2 to 20 nucleotides (FIG. 17G ). PCR artifacts were ruled out using independently produced long-read RNAseq data sets derived from oligo-dT-primed RNA from whole SAD brain and SAD temporal lobe, which yielded non-classical variants with similar IEJs (FIG. 17H ). - Non-classical variants of APP were analyzed in genomic DNA from populations of neuronal nuclei.
- High-stringency amplification using the APP primers described above was pursued on thoroughly RNased DNA obtained from sets of 20 neuronal nuclei from both normal and SAD brains (
FIG. 18A ). PCR of nuclear genomic DNA generated clear bands that were similar in size to non-classical variants from RNA-derived RT-PCR products (FIG. 18B , ˜100-2,300 bp). Interrogation of a second AD related gene, Presenilin 1 (PSEN1), did not produce products from genomic DNA (FIG. 18B ; 94 Kb). Cloning and Sanger sequencing of these genomic DNA products revealed a range of genomic cDNAs (gencDNAs) showing precise exon::exon junctions, central exon deletions, and IEJs, including some species with sequences identical to the non-classical RNA variants identified (FIG. 18C ). - The presence of APP gencDNA junctions within single neuronal genomes was analyzed using jgISH.
- Briefly, sample preparation and hybridization protocols for RNA-ISH were used according to manufacturer's instruction (ACD, Newark, Calif.) to recognize genomic DNA sequences. Probes used passed multiple specificity requirements involving both positive and negative controls (Table 6). Two jgISH probes were used: one that recognized gencDNAs via the exon16::
exon 17 junction (Ex 16/17), which spans the Ab coding region of APP; and one that recognized IEJ formed betweenexons 3 and 16 (IEJ 3/16), representing one APP variant. All bound probes were enzymatically visualized, appearing as red dots (as indicated by the arrows) of varied diameter. Both sense (1901, 1905) and anti-sense (1903, 1907) jgISH probes produced similar results in RNase treated SAD neuronal nuclei (FIGS. 19A-19B ). By comparison, RNA signals were only detected using the anti-sense probes (FIGS. 19C-19D ); therefore sense probes were exclusively used for genomic DNA detection. The jgISH sense probe signals were eliminated by specific restriction enzyme digestion of genomic DNA that eliminated the sequence recognition site (FIGS. 19E-19J ). -
TABLE 6 List of jgISH positive control, negative control, and experimental probes Junction Target Sample Type Probes Ex DNA Human nuclei + RNase Exp Sense 16/17 Exp Anti-sense Human nuclei +RNase +/− Neg Sense restriction enzyme (MluCl) Sense WT mouse nuclei +RNase Neg Sense Mouse nuclei +RNase + Pos Sense Ex 16/17 DNA concatamer RNA Human tissue Neg Sense Pos Anti-sense IEJ 3/16 DNA Human nuclei +RNase Exp Sense Exp Anti-sense Human nuclei +RNase +/− Neg Sense restriction enzyme (PSTI & MslI) Sense WT Mouse nuclei +RNase Neg Sense RNA Human tissue Neg Sense Pos Anti-sense Exp = Experimental, Neg = negative control, Pos = positive control - This example shows that the jgISH protocol detected specific genomic junctions without polymerase dependent template amplification. Moreover, use of Ex16/17 and
IEJ 3/16 probes identified the mosaic presence of these gencDNA sequences in neuronal nuclei. - Non-classical gencDNA variants of APP were analyzed using multiple independent reactions on neuronal populations from brains (
FIG. 20A ), utilizing a DNA polymerase with 100× higher fidelity compared to native Taq (Invitrogen, Platinum SuperFi DNA Polymerase). The resulting samples were pooled for library preparation to enable SMRT CCS of single DNA molecules. SMRT libraries yielded high-certainty consensus calling (20 CCS subreads with 99.9999% accuracy, median Phred score of 93). The number of unique sequences included 2,980 sequences. These included 21 different IEJs identified in neuronal nuclei of 2 SAD brains (FIGS. 20B-20E ) and 858 unique sequences including 11 IEJs in neuronal nuclei of 1 non-diseased brain. SeeFIG. 18C . GencDNAs of the canonical neuronal splice variant,APP 751, were also identified in both SAD and non-diseased datasets. SNVs, and insertions and deletions (INDELs) also occurred within APP gencDNAs of both SAD and non-diseased brain (FIGS. 20E-20F ). - GencDNA forms of mosaic neuronal recombination for potential relevance to SAD were assessed. Ten different SNVs in gencDNA variants from SAD but not non-diseased neurons were identified that were identical to pathogenic FAD APP mutations, including the Indiana mutation (
FIG. 20E andFIG. 21A ). Relationships of identified gencDNA variants to SAD were analyzed by comparing non-diseased and SAD neurons using jgISH. - Two gencDNA junctions,
Ex 16/17 andIEJ 3/16, were examined in neurons from clinically and neuropathologically verified SAD brains (See Table 5) and compared to 6 non-diseased brains (FIGS. 21B-21I ). The number of red foci in AD neurons was 3-5 fold higher than in non-diseased neurons. Rare foci were observed in non-neuronal (NeuN-negative) nuclei that were not statistically significant between cells from SAD and non-diseased brain, despite being from the same brains that had revealed disease-related changes in neurons. Recombination of APP at bothEx 16/17 andIEJ 3/16 was present in the J20 neurons (FIGS. 22A-22G ). - The human transgene and non-classical variants of APP were analyzed. Predominant signals within neuronal nuclei, contrasting with low levels in non-neuronal nuclei from the same animals as well as wild-type controls were observed. See
FIGS. 22A-22G . jgISH analyses of J20 neurons aged 177 vs. 829 days identified age-related increases inEx 16/17 foci sizes in neurons (FIGS. 23A-23C ). The size of foci reflected increased DNA copy number, as demonstrated by control experiments in which retroviral-mediated insertion of DNA target sequences with increasing 16/17 copies allowed semi-quantitation of jgISH foci sizes relative to target copy number (FIGS. 23D-23F ). - These results support neuronal gene transcription in generating gencDNAs.
- Co-localization of APP c-terminal and Aβ plaques was determined in human tissue sections of subjects with Alzheimer's disease. Referring to
FIG. 24 , Aβ plaques (“Aβ”) were stained using an antibody recognizing Aβ peptide as seen in green in the first panel from the left. APP c-terminal (“c-terminal”) was stained using an antibody recognizing APP c-terminal as seen in red in arrows in the second panel from the left. DAPI (“DAPI”) was used to stain the nuclei as seen in blue in the fourth panel from the left. When images of Aβ and APP c-terminal were merged (“merge”) as seen in the arrows in the third panel from the left, Aβ and APP c-terminal were found to be co-localized. - The data shows that non-classical variants of APP identified are present in Aβ plaques and are involved in Aβ plaque formation.
- While preferred embodiments of the present invention have been shown and described herein, it will be obvious to those skilled in the art that such embodiments are provided by way of example only. Numerous variations, changes, and substitutions will now occur to those skilled in the art without departing from the invention. It should be understood that various alternatives to the embodiments of the invention described herein may be employed in practicing the invention. It is intended that the following claims define the scope of the invention and that methods and structures within the scope of these claims and their equivalents be covered thereby.
Claims (132)
Priority Applications (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US16/610,293 US20200080154A1 (en) | 2017-05-02 | 2018-05-01 | Methods of diagnosing and treating alzheimer's disease |
Applications Claiming Priority (3)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US201762500270P | 2017-05-02 | 2017-05-02 | |
US16/610,293 US20200080154A1 (en) | 2017-05-02 | 2018-05-01 | Methods of diagnosing and treating alzheimer's disease |
PCT/US2018/030520 WO2018204408A1 (en) | 2017-05-02 | 2018-05-01 | Methods of diagnosing and treating alzheimer's disease |
Publications (1)
Publication Number | Publication Date |
---|---|
US20200080154A1 true US20200080154A1 (en) | 2020-03-12 |
Family
ID=64016942
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
US16/610,293 Pending US20200080154A1 (en) | 2017-05-02 | 2018-05-01 | Methods of diagnosing and treating alzheimer's disease |
Country Status (2)
Country | Link |
---|---|
US (1) | US20200080154A1 (en) |
WO (1) | WO2018204408A1 (en) |
Families Citing this family (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
EP3810142A4 (en) * | 2018-06-20 | 2022-03-30 | Sanford Burnham Prebys Medical Discovery Institute | Anti-retroviral therapies and reverse transcriptase inhibitors for treatment of alzheimer's disease |
CN116348487A (en) | 2020-07-23 | 2023-06-27 | 欧萨尔普罗席纳有限公司 | Anti-amyloid beta antibodies |
KR102364890B1 (en) * | 2020-10-12 | 2022-02-18 | 경북대학교 산학협력단 | Expression cassette and vector with genes related alzheimer's disease and transgenic human pluripotent stem cell line made from it and alzheimer's diseae organoid and neuron model differentiated from this cell line |
CN114634982A (en) * | 2020-12-15 | 2022-06-17 | 广州市基准医疗有限责任公司 | Method for detecting polynucleotide variation |
Citations (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20040167161A1 (en) * | 2002-11-29 | 2004-08-26 | Masako Nozaki | Method of treating or inhibiting the development of brain inflammation and sepsis |
WO2005003387A2 (en) * | 2003-06-30 | 2005-01-13 | Stichting Katholieke Universiteit, University Medical Centre Nijmegen | Specific method of prostate cancer detection based on pca3 gene, and kits therefor |
US20060270841A1 (en) * | 2002-02-27 | 2006-11-30 | Espeseth Amy S | Assays to monitor amyloid precursor protein processing |
US20140235473A1 (en) * | 2011-06-24 | 2014-08-21 | Markus Otto | Diagnosis and/or Prognosis of Parkinson's Disease Dementia |
Family Cites Families (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2009105572A2 (en) * | 2008-02-19 | 2009-08-27 | Edunn Biotechonology Inc. | Antisense modulation of amyloid beta protein expression |
UA108363C2 (en) * | 2009-10-08 | 2015-04-27 | IMINOTIADIASIADIOXIDE OXIDES AS BACE INHIBITORS, COMPOSITIONS THEREOF AND THEIR APPLICATIONS | |
CA2874762A1 (en) * | 2012-05-31 | 2013-12-05 | Ibis Biosciences, Inc. | Universal random access detection of nucleic acids |
-
2018
- 2018-05-01 WO PCT/US2018/030520 patent/WO2018204408A1/en active Application Filing
- 2018-05-01 US US16/610,293 patent/US20200080154A1/en active Pending
Patent Citations (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20060270841A1 (en) * | 2002-02-27 | 2006-11-30 | Espeseth Amy S | Assays to monitor amyloid precursor protein processing |
US20040167161A1 (en) * | 2002-11-29 | 2004-08-26 | Masako Nozaki | Method of treating or inhibiting the development of brain inflammation and sepsis |
WO2005003387A2 (en) * | 2003-06-30 | 2005-01-13 | Stichting Katholieke Universiteit, University Medical Centre Nijmegen | Specific method of prostate cancer detection based on pca3 gene, and kits therefor |
US20140235473A1 (en) * | 2011-06-24 | 2014-08-21 | Markus Otto | Diagnosis and/or Prognosis of Parkinson's Disease Dementia |
Non-Patent Citations (12)
Title |
---|
Anand et al., A review on cholinesterase inhibitors for Alzheimer's disease, Archives of Pharmacal Research, volume 36, pages 375-399. (Year: 2013) * |
Di Fede et al., A recessive mutation in the APP gene with dominant-negative effect on amyloidogenesis, Science, volume 323, pages 1473-1477. (Year: 2009) * |
Ghidoni et al., Novel T719P AbPP mutation unbalances the relative proportion of Amyloid-b peptides, Journal of Alzheimer's Disease, volume 18, pages 295-303. (Year: 2009) * |
Kaj Blennow, A review of fluid biomarkers for Alzheimer's disease: moving from CSF to blood, Neurology and Therapy, volume 6, supplement 1, pages S15-S24. (Year: 2017) * |
Kim et al., APP gene copy number changes reflect exogenous contamination, Nature, volume 584, pages E20-E33. (Year: 2020) * |
König et al., Identification and differential expression of a novel alternative splice isoform of the betaA4 amyloid precursor protein (APP) mRNA in leukocytes and brain microglial cells, The Journal of Biological Chemistry, volume 267, pages 10804-10809. (Year: 1992) * |
Lee et al., Mosaic APP gene recombination in Alzheimer's disease - What's next? Journal of Experimental Neuroscience, volume 13, pages 1-4. (Year: 2019) * |
Lee et al., Somatic APP gene recombination in Alzheimer's disease and normal neurons, Nature, volume 563, pages 639-645. (Year: 2018) * |
Nicolas et al., Screening of dementia genes by whole-exome sequencing in early-onset Alzheimer disease: input and lessons, European Journal of Human Genetics, volume 24, pages 710-716. (Year: 2016) * |
Parcerisas et al., Somatic signature of brain-specific single nucleotide variations in sporadic Alzheimer's disease, Journal of Alzheimer's Disease, volume 42, pages 1357-1382. (Year: 2014) * |
Shi et al., Long-read sequencing and de novo assembly of a Chinese genome, Nature Communications, volume 7:12065, pages 1-10. (Year: 2015) * |
Tang et al., Identification of a novel alternative splicing isoforms of human amyloid precursor protein, APP639, European Journal of Neuroscience, volume 18, pages 102-108. (Year: 2003) * |
Also Published As
Publication number | Publication date |
---|---|
WO2018204408A1 (en) | 2018-11-08 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
Lee et al. | Somatic APP gene recombination in Alzheimer’s disease and normal neurons | |
US20210267995A1 (en) | Anti-retroviral therapies and reverse transcriptase inhibitors for treatment of alzheimer's disease | |
Swarup et al. | Identification of evolutionarily conserved gene networks mediating neurodegenerative dementia | |
US20200080154A1 (en) | Methods of diagnosing and treating alzheimer's disease | |
Colin et al. | Loss-of-function mutations in WDR73 are responsible for microcephaly and steroid-resistant nephrotic syndrome: Galloway-Mowat syndrome | |
US20180258491A1 (en) | Biomarkers associated with brm inhibition | |
JP2019512489A (en) | MicroRNA and method of using the same | |
AU2010291933A1 (en) | Micro-RNA, autoantibody and protein markers for diagnosis of neuronal injury | |
US9260755B2 (en) | Compositions and methods for characterizing and treating muscular dystrophy | |
CN107250378B (en) | Method for diagnosis, prevention and treatment of brain metastasis of cancer, and drug delivery system for crossing blood brain barrier | |
US20130210663A1 (en) | Methods and compounds for the diagnosis and treatment of cancer | |
Sabino et al. | Epigenetic reprogramming by TET enzymes impacts co-transcriptional R-loops | |
CA3211847A1 (en) | Circular rnas for diagnosis of depression and prediction of response to antidepressant treatment | |
CA2980393A1 (en) | Methods for diagnosing and treating follicular lymphoma | |
KR102547432B1 (en) | Pharmaceutical Composition Comprising Modulator of TUT4/7 Expression | |
KR20200145930A (en) | Markers for diagnosing epilepsy and uses thereof | |
Gasperini et al. | The piRNA pathway sustains adult neurogenesis by reducing protein synthesis and cellular senescence | |
US20180087098A1 (en) | Methods for detecting rapidly processed introns to evaulate allelic expression | |
Alessandrini et al. | ALS-Associated TDP-43 Dysfunction Compromises UPF1-Dependent mRNA Metabolism Pathways Including Alternative Polyadenylation and 3'UTR Length | |
US10590488B2 (en) | Recurrent gene fusions in cutaneous CD30-positive lymphoproliferative disorders | |
CA3070462A1 (en) | Methods and compositions for viral nano-fish | |
US20240353397A1 (en) | Markers of resistance and disease tolerance and uses thereof | |
KR102256859B1 (en) | Composition for diagnosing Alzheimer's disease | |
US20210254161A1 (en) | Method for determining decrease in functions of hippocampus by using correlation between micro rna and nmda receptor, method for inhibiting decrease in functions, and method for screening for inhibitors of decrease in functions | |
Kuriakose et al. | Upstream regulation of microRNA-9 through a complex cellular machinery during neurogenesis |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
AS | Assignment |
Owner name: SANFORD BURNHAM PREBYS MEDICAL DISCOVERY INSTITUTE, CALIFORNIA Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:CHUN, JEROLD;LEE, MING-HSIANG;REEL/FRAME:053437/0706 Effective date: 20191218 |
|
STPP | Information on status: patent application and granting procedure in general |
Free format text: APPLICATION DISPATCHED FROM PREEXAM, NOT YET DOCKETED |
|
STPP | Information on status: patent application and granting procedure in general |
Free format text: NON FINAL ACTION MAILED |
|
STPP | Information on status: patent application and granting procedure in general |
Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER |
|
STPP | Information on status: patent application and granting procedure in general |
Free format text: NON FINAL ACTION MAILED |
|
STPP | Information on status: patent application and granting procedure in general |
Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER |
|
STPP | Information on status: patent application and granting procedure in general |
Free format text: FINAL REJECTION MAILED |
|
STPP | Information on status: patent application and granting procedure in general |
Free format text: NON FINAL ACTION MAILED |
|
STPP | Information on status: patent application and granting procedure in general |
Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER |
|
STPP | Information on status: patent application and granting procedure in general |
Free format text: FINAL REJECTION MAILED |
|
STPP | Information on status: patent application and granting procedure in general |
Free format text: RESPONSE AFTER FINAL ACTION FORWARDED TO EXAMINER |
|
STPP | Information on status: patent application and granting procedure in general |
Free format text: ADVISORY ACTION MAILED |
|
STPP | Information on status: patent application and granting procedure in general |
Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION |
|
STPP | Information on status: patent application and granting procedure in general |
Free format text: NON FINAL ACTION MAILED |